1
|
Choi SYC, Ribeiro CF, Wang Y, Loda M, Plymate SR, Uo T. Druggable Metabolic Vulnerabilities Are Exposed and Masked during Progression to Castration Resistant Prostate Cancer. Biomolecules 2022; 12:1590. [PMID: 36358940 PMCID: PMC9687810 DOI: 10.3390/biom12111590] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 08/27/2023] Open
Abstract
There is an urgent need for exploring new actionable targets other than androgen receptor to improve outcome from lethal castration-resistant prostate cancer. Tumor metabolism has reemerged as a hallmark of cancer that drives and supports oncogenesis. In this regard, it is important to understand the relationship between distinctive metabolic features, androgen receptor signaling, genetic drivers in prostate cancer, and the tumor microenvironment (symbiotic and competitive metabolic interactions) to identify metabolic vulnerabilities. We explore the links between metabolism and gene regulation, and thus the unique metabolic signatures that define the malignant phenotypes at given stages of prostate tumor progression. We also provide an overview of current metabolism-based pharmacological strategies to be developed or repurposed for metabolism-based therapeutics for castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Stephen Y. C. Choi
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Caroline Fidalgo Ribeiro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10021, USA
- New York Genome Center, New York, NY 10013, USA
| | - Stephen R. Plymate
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
- Geriatrics Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA 98108, USA
| | - Takuma Uo
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, 850 Republican St., Seattle, WA 98109, USA
| |
Collapse
|
2
|
Surcel C, Kretschmer A, Mirvald C, Sinescu I, Heidegger I, Tsaur I. Molecular Mechanisms Related with Oligometastatic Prostate Cancer-Is It Just a Matter of Numbers? Cancers (Basel) 2022; 14:cancers14030766. [PMID: 35159033 PMCID: PMC8833728 DOI: 10.3390/cancers14030766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023] Open
Abstract
During the last decade, the body of knowledge regarding the oligometastatic state has increased exponentially. Several molecular frameworks have been established, aiding our understanding of metastatic spread caused by genetically unstable cells that adapt to a tissue environment which is distant from the primary tumor. In the current narrative review, we provide an overview of the current treatment landscape of oligometastatic cancer, focusing on the current biomarkers used in the identification of true oligometastatic disease and highlighting the impact of molecular imaging on stage shift in different scenarios. Finally, we address current and future directions regarding the use of genetic and epigenetic targeting treatments in oligometastatic prostate cancer.
Collapse
Affiliation(s)
- Cristian Surcel
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 00238 Bucharest, Romania; (C.M.); (I.S.)
- Correspondence:
| | | | - Cristian Mirvald
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 00238 Bucharest, Romania; (C.M.); (I.S.)
| | - Ioanel Sinescu
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 00238 Bucharest, Romania; (C.M.); (I.S.)
| | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, 6020 Innsbruck, Austria;
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany;
| |
Collapse
|
3
|
Elgazzar AH, Sarikaya I. Basis of Therapeutic Nuclear Medicine. THE PATHOPHYSIOLOGIC BASIS OF NUCLEAR MEDICINE 2022:569-594. [DOI: 10.1007/978-3-030-96252-4_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Filippi L, Chiaravalloti A, Basile P, Schillaci O, Bagni O. Molecular and metabolic imaging of castration-resistant prostate cancer: state of art and future prospects. Curr Mol Med 2021; 22:25-36. [PMID: 33573553 DOI: 10.2174/1566524021666210211112423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/23/2020] [Accepted: 02/02/2021] [Indexed: 11/22/2022]
Abstract
Prostate cancer (PCa) represents the most common tumor in male and one of the most relevant causes of death in Western countries. Androgen deprivation therapy (ADT) constitutes a widely used approach in advanced PCa. When PCa progresses in spite of ADT and castrate levels of testosterone, the severe clinical condition termed as metastatic castration-resistant prostate cancer (mCRPC) takes place. The only approach to mCRPC has been represented by chemotherapy with taxanes for many years. Nevertheless, recently introduced treatments such as 2nd generation antiandrogens (i.e. enzalutamide and abiraterone), cell immunotherapy with sipuleucel-T or targeted alpha therapy with 223Ra-dichloride, have dramatically changed mCRPC prognosis. These novel therapies call for an unmet need for imaging biomarkers suitable for patients' pre-treatment stratification and response assessment. In this scenario, nuclear medicine can provide several metabolic and molecular probes for investigating pathological processes at a cellular and sub-cellular level. The aim of this paper is to review the most relevant findings of the literature published to date on this topic, giving particular emphasis to the pros and cons of each tracer and also covering future prospects for defining personalized therapeutic approaches.
Collapse
Affiliation(s)
- Luca Filippi
- Nuclear Medicine Department, "Santa Maria Goretti" Hospital, via Canova, 04100, Latina. Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University Tor Vergata, Viale Oxford 81, 00133, Rome. Italy
| | - Pietro Basile
- Nuclear Medicine Department, "Santa Maria Goretti" Hospital, via Canova, 04100, Latina. Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Viale Oxford 81, 00133, Rome. Italy
| | - Oreste Bagni
- Nuclear Medicine Department, "Santa Maria Goretti" Hospital, via Canova, 04100, Latina. Italy
| |
Collapse
|
5
|
Salvage Pelvic Lymph Node Dissection and Current State of Imaging for Recurrent Prostate Cancer: Does a Standard Exist? Curr Urol Rep 2020; 21:62. [PMID: 33159608 DOI: 10.1007/s11934-020-01011-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW We aim to evaluate the efficacy of salvage lymph node dissection (SLND) for nodal recurrent prostate cancer after primary treatment. We also provide a review of the diagnostic performance of next-generation sequencing (next-generation imaging (NGI)) radiotracers in the salvage setting. RECENT FINDINGS Most studies evaluating SLND include a heterogeneous population with a small sample size and are retrospective in design. The 5-year clinical recurrence-free and cancer-specific survival following SLND are 26-52% and 57-89%, respectively, among prospective studies. NGI improves accuracy in detecting nodal recurrence compared to conventional CT, with PMSA PET-CT showing the most promise. However, limited studies exist comparing imaging modalities and performance is variable at low PSA values. SLND is a promising treatment option, but more prospective data are needed to determine the ideal surgical candidate and long-term oncologic outcomes. More studies comparing different NGI are needed to determine the best imaging modality in patients who may be candidates for salvage treatment.
Collapse
|
6
|
Mankoff DA, Pantel AR, Viswanath V, Karp JS. Advances in PET Diagnostics for Guiding Targeted Cancer Therapy and Studying In Vivo Cancer Biology. CURRENT PATHOBIOLOGY REPORTS 2019; 7:97-108. [PMID: 37092138 PMCID: PMC10117535 DOI: 10.1007/s40139-019-00202-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose of the Review We present an overview of recent advances in positron emission tomography (PET) diagnostics as applied to the study of cancer, specifically as a tool to study in vivo cancer biology and to direct targeted cancer therapy. The review is directed to translational and clinical cancer investigators who may not be familiar with these applications of PET cancer diagnostics, but whose research might benefit from these advancing tools. Recent Findings We highlight recent advances in 3 areas: (1) the translation of PET imaging cancer biomarkers to clinical trials; (2) methods for measuring cancer metabolism in vivo in patients; and (3) advances in PET instrumentation, including total-body PET, that enable new methodologies. We emphasize approaches that have been translated to human studies. Summary PET imaging methodology enables unique in vivo cancer diagnostics that go beyond cancer detection and staging, providing an improved ability to guide cancer treatment and an increased understanding of in vivo human cancer biology.
Collapse
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Austin R Pantel
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Varsha Viswanath
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Joel S Karp
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
7
|
Theranostics for Advanced Prostate Cancer: Current Indications and Future Developments. Eur Urol Oncol 2019; 2:152-162. [PMID: 31017091 DOI: 10.1016/j.euo.2019.01.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022]
Abstract
CONTEXT Advanced prostate cancer (PCa) is a prominent cause of cancer death in men; positron emission tomography (PET) imaging may play a relevant role in detecting metastases and thus allowing a more tailored therapy in these patients. Radioligand therapy (RLT) may also gain relevance as a treatment strategy in advanced disease. OBJECTIVE The aim of this review is to highlight how the recently developed theranostic processes may become a part of both the available diagnostic and the therapy arsenal in advanced PCa patients. EVIDENCE ACQUISITION An expert panel of nuclear medicine physicians and a urologist, highly experienced in the fields of radionuclide imaging and RLT in advanced PCa, performed a nonsystematic review of the current indications, performance, limitations, and potential future developments of the currently available options in PCa theranostics. EVIDENCE SYNTHESIS Among PET radiotracers, prostate-specific membrane antigen (PSMA)-based compounds in advanced PCa are the focus of a continuously growing interest, mostly due to their potential relevance as theranostic agents. The impact of PSMA-based PET/computed tomography imaging on treatment strategies and prognosis is promising, but still not unquestionably clear. Potential applications may include a role as a gatekeeper to PSMA-directed RLT, as well as monitoring the spread of systemic disease. Currently, initial results seem to substantiate the role of PSMA-directed RLT in terms of feasibility and efficacy. CONCLUSIONS PSMA is a promising molecule for both imaging and therapy in advanced PCa patients; nevertheless, further studies are needed to investigate its role and to determine the impact of its side effects and its overall strategy outcome. PATIENT SUMMARY Prostate-specific membrane antigen (PSMA), a protein, is highly expressed on prostate cancer cells. The possibility to perform diagnostic imaging and subsequently administer therapies by the means of the same molecule is called "theranostics". In patients with advanced prostate cancer, PSMA might have a role in detecting disease spread through both positron emission tomography and single-photon emission computed tomography imaging, while treating prostate cancer systemic localizations with radioligand therapy. Further studies are needed to better determine patients' risks and benefits of these therapeutic approaches.
Collapse
|
8
|
Mortensen MA, Vilstrup MH, Poulsen MH, Gerke O, Høilund-Carlsen PF, Lund L. A prospective study on dual time 18F-FDG-PET/CT in high-risk prostate cancer patients. BMC Res Notes 2018; 11:871. [PMID: 30526642 PMCID: PMC6286604 DOI: 10.1186/s13104-018-3985-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/04/2018] [Indexed: 11/26/2022] Open
Abstract
Objective This proof of concept study investigated whether dual time point FDG-PET/CT with image acquisition after 1 and 3 h could be useful in preoperative staging of patients undergoing robot-assisted radical prostatectomy and extended pelvic lymph node dissection for high-risk prostate cancer. Results Twenty patients with high-risk prostate cancer underwent dual time point FDG-PET/CT before undergoing surgery. Histologically confirmed lymph node metastases were found in 9/20 (45%). A median of 19 (range 10–41; n = 434) lymph nodes were removed per patient. Pelvic lymph nodes with detectable FDG uptake were seen in two patients only, but the FDG-avid lesion on PET did not correspond with pathological findings in either patient. We found a significant increase in maximal standardized uptake value of the prostate of around 30% between early and late imaging. We found no correlation between clinical findings after radical prostatectomy and PET measurements.
Collapse
Affiliation(s)
- Mike Allan Mortensen
- Department of Urology, Odense University Hospital, Odense, Denmark. .,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Mie Holm Vilstrup
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Mads Hvid Poulsen
- Department of Urology, Odense University Hospital, Odense, Denmark.,Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, Odense, Denmark
| | - Oke Gerke
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Poul Flemming Høilund-Carlsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
9
|
Pantel AR, Ackerman D, Lee SC, Mankoff DA, Gade TP. Imaging Cancer Metabolism: Underlying Biology and Emerging Strategies. J Nucl Med 2018; 59:1340-1349. [PMID: 30042161 PMCID: PMC6126440 DOI: 10.2967/jnumed.117.199869] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 06/18/2018] [Indexed: 12/22/2022] Open
Abstract
Dysregulated cellular metabolism is a characteristic feature of malignancy that has been exploited for both imaging and targeted therapy. With regard to imaging, deranged glucose metabolism has been leveraged using 18F-FDG PET. Metabolic imaging with 18F-FDG, however, probes only the early steps of glycolysis; the complexities of metabolism beyond these early steps in this single pathway are not directly captured. New imaging technologies-both PET with novel radiotracers and MR-based methods-provide unique opportunities to investigate other aspects of cellular metabolism and expand the metabolic imaging armamentarium. This review will discuss the underlying biology of metabolic dysregulation in cancer, focusing on glucose, glutamine, and acetate metabolism. Novel imaging strategies will be discussed within this biologic framework, highlighting particular strengths and limitations of each technique. Emphasis is placed on the role that combining modalities will play in enabling multiparametric imaging to fully characterize tumor biology to better inform treatment.
Collapse
Affiliation(s)
- Austin R Pantel
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel Ackerman
- Penn Image-Guided Interventions Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Seung-Cheol Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A Mankoff
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Terence P Gade
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Image-Guided Interventions Laboratory, Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania; and
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Mankoff DA, Katz SI. PET imaging for assessing tumor response to therapy. J Surg Oncol 2018; 118:362-373. [PMID: 29938396 DOI: 10.1002/jso.25114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Abstract
Positron emission tomography (PET) is a radioisotope imaging technique capable of quantifying the regional distribution of molecular imaging probes targeted to biochemical pathways and processes allowing direct measurement of biochemical changes induced by cancer therapy, including the activity of targeted growth pathways and cellular populations. In this manuscript, we review the underlying principles of PET imaging, choices for PET radiopharmaceuticals, methods for tumor analysis and PET applications for cancer therapy response assessment including potential future directions.
Collapse
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sharyn I Katz
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Imaging the High-risk Prostate Cancer Patient: Current and Future Approaches to Staging. Urology 2018; 116:3-12. [DOI: 10.1016/j.urology.2017.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 11/28/2017] [Accepted: 12/01/2017] [Indexed: 11/24/2022]
|
12
|
Spick C, Herrmann K, Czernin J. Evaluation of Prostate Cancer with 11C-Acetate PET/CT. J Nucl Med 2017; 57:30S-37S. [PMID: 27694168 DOI: 10.2967/jnumed.115.169599] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/10/2016] [Indexed: 11/16/2022] Open
Abstract
In this article, we will first describe the metabolic fate of 11C-acetate; then discuss its biodistribution in health and disease; and subsequently focus on its key clinical applications, the detection and localization of prostate cancer tissue in patients with primary or recurrent disease. Finally, we will discuss the potential role of 11C-acetate in the context of other prostate cancer imaging probes and non-radionuclide-based imaging approaches.
Collapse
Affiliation(s)
- Claudio Spick
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Ken Herrmann
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Johannes Czernin
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
13
|
Ballas LK, de Castro Abreu AL, Quinn DI. What Medical, Urologic, and Radiation Oncologists Want from Molecular Imaging of Prostate Cancer. J Nucl Med 2017; 57:6S-12S. [PMID: 27694176 DOI: 10.2967/jnumed.115.170142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 12/25/2022] Open
Abstract
As molecular imaging better delineates the state of prostate cancer, clinical management will evolve. The currently licensed imaging modalities are limited by lack of specificity or sensitivity for the extent of cancer and for predicting outcome in response to therapy. Clinicians want molecular imaging that-by being more reliable in tailoring treatment and monitoring response for each patient-will become a key facet of precision medicine, surgery, and radiation therapy. Identifying patients who are candidates for specific or novel treatments is important, but equally important is the finding that a given patient may not be a good candidate for single-modality therapy. This article presents prostate cancer scenarios in which managing clinicians would welcome molecular imaging innovations to help with decision making. The potential role of newer techniques that may help fill this wish list is discussed.
Collapse
Affiliation(s)
- Leslie K Ballas
- Department of Radiation Oncology, Keck School of Medicine at USC, USC Norris Comprehensive Cancer Center and Hospital, Los Angeles, California
| | - Andre Luis de Castro Abreu
- Department of Urology, Keck School of Medicine at USC, USC Norris Comprehensive Cancer Center and Hospital, Los Angeles, California; and
| | - David I Quinn
- Division of Medical Oncology, Department of Medicine, Keck School of Medicine at USC, USC Norris Comprehensive Cancer Center and Hospital, Los Angeles, California
| |
Collapse
|
14
|
Management of metastatic castration-resistant prostate cancer: A focus on radium-223: Opinions and suggestions from an expert multidisciplinary panel. Crit Rev Oncol Hematol 2017; 113:43-51. [PMID: 28427521 DOI: 10.1016/j.critrevonc.2017.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 01/22/2023] Open
Abstract
Radium-223, a calcium mimetic bone-seeking radionuclide that selectively targets bone metastases with alpha particles, is approved for the treatment of men with metastatic castration-resistant prostate cancer (mCRPC) and symptomatic bone metastases. In patients with mCRPC, treatment with radium-223 has been associated with survival benefit, regardless of prior docetaxel use, and also has a positive impact on symptomatic skeletal events and quality of life. Radium-223 is best suited for patients with symptomatic mCRPC and bone-predominant disease and no visceral metastases, and may lead to better outcomes when given early in the course of the disease. An expert multidisciplinary panel convened in Milan, Italy to review the current best-evidence literature on radium-223 and to convey their personal expertise with the use of radium-223 and identify possible strategies for best practice. This article summarizes the best available evidence for the use of radium-223, discusses the essential role of the multidisciplinary team in delivering effective treatment for mCRPC, clarifies pre- and post-treatment evaluation and monitoring, and outlines future scenarios for radium-223 in the treatment of men with MCRPC.
Collapse
|
15
|
Almeida FD, Yen CK, Scholz MC, Lam RY, Turner J, Bans LL, Lipson R. Performance characteristics and relationship of PSA value/kinetics on carbon-11 acetate PET/CT imaging in biochemical relapse of prostate cancer. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2017; 7:1-11. [PMID: 28123863 PMCID: PMC5259584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/09/2016] [Indexed: 06/06/2023]
Abstract
An elevated serum prostate-specific antigen (PSA) level alone cannot distinguish between local-regional recurrences and distant metastases after treatment with curative intent. With available salvage treatments, it has become important to localize the site of recurrence. 11C-Acetate PET/CT was performed in patients with rising PSA, with statistical analysis of detection rates, sites/location of detection, PSA kinetics and comparison with other tracers (FDG and Choline). Correlation to biopsy, subsequent imaging and PSA response to focal treatment was also performed. 88% (637) of 721 11C-Acetate PET/CT scans performed were positive. There was a statistically significant difference in PSA values between the positive and negative scans (P < 0.001 for mean difference) with the percentage of positive scans and PSA having a positive correlation. A PSA of 1.09 ng/mL was found to be an optimal cutoff. PSAdT was significantly correlated with a positive scan only when the PSA was < 1.0 ng/mL. For this subgroup, a PSAdT of < 3.8 months appeared significant (P < 0.05) as an optimal cutoff point. 11C-Acetate PET/CT demonstrates a high detection rate for the site of recurrence/metastasis in biochemical relapsed prostate cancer (88% overall detection rate, PPV 90.8%). This analysis suggests an optimal PSA threshold of > 1.09 ng/mL or a PSAdT of < 3.8 months when the PSA is below 1.0 ng/mL as independent predictors of positive findings.
Collapse
Affiliation(s)
| | | | - Mark C Scholz
- Prostate Oncology Specialists Marina Del Rey, CA, USA
| | - Richard Y Lam
- Prostate Oncology Specialists Marina Del Rey, CA, USA
| | | | | | | |
Collapse
|
16
|
Fontana F, Ge X, Su X, Hathi D, Xiang J, Cenci S, Civitelli R, Shoghi KI, Akers WJ, D'avignon A, Weilbaecher KN, Shokeen M. Evaluating Acetate Metabolism for Imaging and Targeting in Multiple Myeloma. Clin Cancer Res 2016; 23:416-429. [PMID: 27486177 DOI: 10.1158/1078-0432.ccr-15-2134] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE We hypothesized that in multiple myeloma cells (MMC), high membrane biosynthesis will induce acetate uptake in vitro and in vivo Here, we studied acetate metabolism and targeting in MMC in vitro and tested the efficacy of 11C-acetate-positron emission tomography (PET) to detect and quantitatively image myeloma treatment response in vivo EXPERIMENTAL DESIGN: Acetate fate tracking using 13C-edited-1H NMR (nuclear magnetic resonance) was performed to study in vitro acetate uptake and metabolism in MMC. Effects of pharmacological modulation of acetate transport or acetate incorporation into lipids on MMC cell survival and viability were assessed. Preclinical mouse MM models of subcutaneous and bone tumors were evaluated using 11C-acetate-PET/CT imaging and tissue biodistribution. RESULTS In vitro, NMR showed significant uptake of acetate by MMC and acetate incorporation into intracellular metabolites and membrane lipids. Inhibition of lipid synthesis and acetate transport was toxic to MMC, while sparing resident bone cells or normal B cells. In vivo, 11C-acetate uptake by PET imaging was significantly enhanced in subcutaneous and bone MMC tumors compared with unaffected bone or muscle tissue. Likewise, 11C-acetate uptake was significantly reduced in MM tumors after treatment. CONCLUSIONS Uptake of acetate from the extracellular environment was enhanced in MMC and was critical to cellular viability. 11C-Acetate-PET detected the presence of myeloma cells in vivo, including uptake in intramedullary bone disease. 11C-Acetate-PET also detected response to therapy in vivo Our data suggested that acetate metabolism and incorporation into lipids was crucial to MM cell biology and that 11C-acetate-PET is a promising imaging modality for MM. Clin Cancer Res; 23(2); 416-29. ©2016 AACR.
Collapse
Affiliation(s)
- Francesca Fontana
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Xia Ge
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Xinming Su
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Deep Hathi
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Jingyu Xiang
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Simone Cenci
- San Raffaele Scientific Institute (SRSI), Division of Genetics and Cell Biology, and Università Vita-Salute San Raffaele, Milano, Italy
| | - Roberto Civitelli
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, Missouri
| | - Kooresh I Shoghi
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Walter J Akers
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Andre D'avignon
- Department of Chemistry, Washington University, St. Louis, Missouri
| | - Katherine N Weilbaecher
- Department of Medicine, Division of Molecular Oncology, Washington University School of Medicine, St. Louis, Missouri.
| | - Monica Shokeen
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
17
|
Farnebo J, Wadelius A, Sandström P, Nilsson S, Jacobsson H, Blomqvist L, Ullén A. Progression-free and overall survival in metastatic castration-resistant prostate cancer treated with abiraterone acetate can be predicted with serial C11-acetate PET/CT. Medicine (Baltimore) 2016; 95:e4308. [PMID: 27495034 PMCID: PMC4979788 DOI: 10.1097/md.0000000000004308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
In this retrospective study, we evaluated the benefit of repeated carbon 11 (C11)-acetate positron emission tomography/computed tomography (PET/CT) to assess response in patients with metastatic castration-resistant prostate cancer (mCRPC) treated with abiraterone acetate (AA).A total of 30 patients with mCRPC were monitored with C11-acetate PET/CT and PSA levels during their treatment with AA. Retrospective evaluation of their response was made after 102 days (median; range 70-155) of treatment. Statistical analyses were employed to detect predictors of progression-free survival (PFS) and overall survival (OS), and potential correlation between serum levels of PSA, standardized uptake values (SUVpeak), and bone lesion index measured from PET were investigated.At follow-up 10 patients exhibited partial response (PR), 10 progressive disease (PD), and 10 stable disease (SD), as assessed by PET/CT. In survival analysis, both PR and PD were significantly associated with PFS and OS. CT response was also associated with OS, but only 19/30 patients demonstrated a lesion meeting target lesion criteria according to RECIST 1.1. No PET/CT baseline characteristic was significantly associated with PFS or OS. A PSA response (reduction in the level by >50%) could also predict PFS and OS. In the subgroup lacking a PSA response, those with PD had significantly shorter OS than those with PR or SD.PFS and OS in patients with mCRPC treated with AA can be predicted from repeated C11-acetate PET/CT. This may be of particular clinical value in patients who do not exhibit a PSA response to treatment.
Collapse
Affiliation(s)
- Jacob Farnebo
- Department of Diagnostic Radiology and Nuclear Medicine, Karolinska University Hospital and Department of Molecular Medicine and Surgery
- Correspondence: Jacob Farnebo, Stavgårdsgatan, Bromma, Sweden (e-mail: )
| | - Agnes Wadelius
- Department of Oncology, Karolinska University Hospital and Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Per Sandström
- Department of Oncology, Karolinska University Hospital and Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Sten Nilsson
- Department of Oncology, Karolinska University Hospital and Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hans Jacobsson
- Department of Diagnostic Radiology and Nuclear Medicine, Karolinska University Hospital and Department of Molecular Medicine and Surgery
| | - Lennart Blomqvist
- Department of Diagnostic Radiology and Nuclear Medicine, Karolinska University Hospital and Department of Molecular Medicine and Surgery
| | - Anders Ullén
- Department of Oncology, Karolinska University Hospital and Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
18
|
Mahajan A, Azad GK, Cook GJ. PET Imaging of Skeletal Metastases and Its Role in Personalizing Further Management. PET Clin 2016; 11:305-318. [PMID: 27321034 DOI: 10.1016/j.cpet.2016.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In oncology, the skeleton is one of the most frequently encountered sites for metastatic disease and thus early detection not only has an impact on an individual patient's management but also on the overall outcome. Multiparametric and multimodal hybrid PET/computed tomography and PET/MR imaging have revolutionized imaging for bone metastases, but irrespective of tumor biology or morphology of the bone lesion it remains unclear which imaging modality is the most clinically relevant to guide individualized cancer care. In this review, we highlight the current clinical challenges of PET imaging in evaluation and quantification of skeletal tumor burden and its impact on personalized cancer management.
Collapse
Affiliation(s)
- Abhishek Mahajan
- Department of Radiodiagnosis, Tata Memorial Hospital, Parel, Mumbai 400012, India
| | - Gurdip Kaur Azad
- Division of Imaging Sciences and Biomedical Engineering, Cancer Imaging Department, King's College London, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK
| | - Gary J Cook
- Division of Imaging Sciences and Biomedical Engineering, Cancer Imaging Department, King's College London, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK; Clinical PET Centre, St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, UK.
| |
Collapse
|
19
|
Jadvar H. Positron emission tomography in imaging evaluation of staging, restaging, treatment response, and prognosis in prostate cancer. Abdom Radiol (NY) 2016; 41:889-98. [PMID: 27193789 DOI: 10.1007/s00261-015-0563-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prostate cancer is a prevalent public health problem worldwide. While imaging has played a major role in this disease, there still remain many challenges and opportunities. Positron emission tomography with various physiologically based radiotracers is fundamentally suited to interrogate this biologically and clinically heterogeneous disease along the course of its natural history. In this article, I review briefly the published evidence for the use of positron emission tomography with 18F-fluorodeoxyglucose, 11C-acetate, and 18F- or 11C-choline in the imaging evaluation of prostate cancer. Although the focus of the article will be on these radiotracers given the accumulated experience with them, but I will also comment on the outlook for the use of other emerging PET radiotracers such as those targeted to the prostate-specific membrane antigen and the amino acid metabolism pathway. It is anticipated that PET will play major role in the evaluation of prostate cancer in the current evidence-based medicine environment. There will also be exciting novel prospects for the use of therapeutic-diagnostic (theransotic) pairs in the management of patients with prostate cancer.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine of USC, University of Southern California, 2250 Alcazar Street, CSC 102, Los Angeles, CA, 90033, USA.
| |
Collapse
|
20
|
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: 43] [Impact Index Per Article: 4.8] [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
|
21
|
Lung Metastasis From Prostate Cancer Revealed by 18F-FDG PET/CT Without Osseous Metastasis on Bone Scan. Clin Nucl Med 2016; 41:392-3. [PMID: 26859201 DOI: 10.1097/rlu.0000000000001107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A 54-year-old man, a case of prostate cancer, underwent radical prostatectomy and hormone therapy. Elevated prostate-specific antigen level developed 7 years later, but pelvic MRI and bone scan revealed negative results. Radiotherapy was performed under the suspicion of local recurrence but in vain. F-FDG PET/CT performed 1 more year later showed 3 FDG-avid lesions in the right lung and mediastinum. Lung and lymph node metastases were proved with video-assisted thoracoscopic surgery. Bone scan remained negative at that time.
Collapse
|
22
|
Azad GK, Taylor B, Rubello D, Colletti PM, Goh V, Cook GJ. Molecular and Functional Imaging of Bone Metastases in Breast and Prostate Cancers: An Overview. Clin Nucl Med 2016; 41:e44-50. [PMID: 26402127 DOI: 10.1097/rlu.0000000000000993] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Our ability to accurately assess the skeleton for metastases in breast and prostate cancers has improved significantly in recent years with hybrid imaging methods. Nevertheless, no consensus has been reached on the best imaging modality for diagnosis and treatment response assessment of skeletal disease. Hybrid SPECT/CT has low false-positive and false-negative rates compared with planar bone scintigraphy (BS) or BS augmented with SPECT in breast and prostate cancers. In breast cancer, 18F-FDG PET is more sensitive and accurate at detecting bone metastases than BS. Currently, little evidence has accrued to support the superiority of 18F-fluoride (18F-NaF) PET in diagnosing osseous metastases or monitoring treatment response in breast cancer when compared with conventional imaging. In prostate cancer, the sensitivities of 18F-NaF PET/CT, 18F-fluorocholine (18F-choline), or 11C-choline PET/CT are equivalent, although 11C-/18F-choline PET/CT scans are more specific. Whole-body MRI, using anatomical sequences complemented by diffusion-weighted MRI, shows early evidence of utility for diagnosis and monitoring therapy response. We review the literature for staging and response assessment in metastatic breast and prostate cancer. While staging accuracy has significantly improved with hybrid imaging, optimal methods for assessing early treatment response have not been determined, and this is an area of active research.
Collapse
|
23
|
Wibmer AG, Burger IA, Sala E, Hricak H, Weber WA, Vargas HA. Molecular Imaging of Prostate Cancer. Radiographics 2015; 36:142-59. [PMID: 26587888 DOI: 10.1148/rg.2016150059] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate cancer is the most common noncutaneous malignancy among men in the Western world. The natural history and clinical course of prostate cancer are markedly diverse, ranging from small indolent intraprostatic lesions to highly aggressive disseminated disease. An understanding of this biologic heterogeneity is considered a necessary requisite in the quest for the adoption of precise and personalized management strategies. Molecular imaging offers the potential for noninvasive assessment of the biologic interactions underpinning prostate carcinogenesis. Currently, numerous molecular imaging probes are in clinical use or undergoing preclinical or clinical evaluation. These probes can be divided into those that image increased cell metabolism, those that target prostate cancer-specific membrane proteins and receptor molecules, and those that bind to the bone matrix adjacent to metastases to bone. The increased metabolism and vascular changes in prostate cancer cells can be evaluated with radiolabeled analogs of choline, acetate, glucose, amino acids, and nucleotides. The androgen receptor, prostate-specific membrane antigen, and gastrin-releasing peptide receptor (ie, bombesin) are overexpressed in prostate cancer and can be targeted by specific radiolabeled imaging probes. Because metastatic prostate cancer cells induce osteoblastic signaling pathways of adjacent bone tissue, bone-seeking radiotracers are sensitive tools for the detection of metastases to bone. Knowledge about the underlying biologic processes responsible for the phenotypes associated with the different stages of prostate cancer allows an appropriate choice of methods and helps avoid pitfalls.
Collapse
Affiliation(s)
- Andreas G Wibmer
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Irene A Burger
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Evis Sala
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Hedvig Hricak
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Wolfgang A Weber
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| | - Hebert Alberto Vargas
- From the Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065 (A.G.W., E.S., H.H., W.A.W., H.A.V.); and Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland (I.A.B.)
| |
Collapse
|
24
|
Bjurlin MA, Rosenkrantz AB, Beltran LS, Raad RA, Taneja SS. Imaging and evaluation of patients with high-risk prostate cancer. Nat Rev Urol 2015; 12:617-28. [PMID: 26481576 DOI: 10.1038/nrurol.2015.242] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Approximately 15% of men with newly diagnosed prostate cancer have high-risk disease. Imaging is critically important for the diagnosis and staging of these patients, and also for the selection of management. While established prostate cancer staging guidelines have increased the appropriate use of imaging, underuse for high-risk prostate cancer remains substantial. Several factors affect the utility of initial diagnostic imaging, including the variable definition of high-risk prostate cancer, variable guideline recommendations, poor accuracy of existing imaging tests, and the difficulty in validating imaging findings. Conventional imaging modalities, including CT and radionuclide bone scan, have been employed for local and metastatic staging, but their performance characteristics have generally been poor. Emerging modalities including multiparametricMRI, positron emission tomography (PET)-CT, and PET-MRI have shown increased diagnostic accuracy and could improve accuracy in staging patients with high-risk prostate cancer.
Collapse
Affiliation(s)
- Marc A Bjurlin
- Division of Urologic Oncology, Department of Urology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Andrew B Rosenkrantz
- Department of Radiology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Luis S Beltran
- Department of Radiology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Roy A Raad
- Department of Radiology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Samir S Taneja
- Division of Urologic Oncology, Department of Urology, New York University Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
25
|
Ouyang Q, Duan Z, Lei J, Jiao G. Comparison of meta-analyses among elastosonography (ES) and positron emission tomography/computed tomography (PET/CT) imaging techniques in the application of prostate cancer diagnosis. Tumour Biol 2015; 37:2999-3007. [PMID: 26415734 DOI: 10.1007/s13277-015-4113-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/20/2015] [Indexed: 10/23/2022] Open
Abstract
The early diagnosis of prostate cancer (PCa) appears to be of vital significance for the provision of appropriate treatment programs. Even though several sophisticated imaging techniques such as positron emission tomography/computed tomography (PET/CT) and elastosonography (ES) have already been developed for PCa diagnosis, the diagnostic accuracy of these imaging techniques is still controversial to some extent. Therefore, a comprehensive meta-analysis in this study was performed to compare the accuracy of various diagnostic imaging methods for PCa, including 11C-choline PET/CT, 11C-acetate PET/CT, 18F-fluorocholine PET/CT, 18F-fluoroglucose PET/CT, transrectal real-time elastosonography (TRTE), and shear-wave elastosonography (SWE). The eligible studies were identified through systematical searching for the literature in electronic databases including PubMed, Cochrane, and Web of Science. On the basis of the fixed-effects model, the pooled sensitivity (SEN), specificity (SPE), and area under the receiver operating characteristics curve (AUC) were calculated to estimate the diagnostic accuracy of 11C-choline PET/CT, 11C-acetate PET/CT, 18F-fluorocholine (FCH) PET/CT, 18F-fluoroglucose (FDG) PET/CT, TRTE, and SWE. All the statistical analyses were conducted with R language Software. The present meta-analysis incorporating a total of 82 studies demonstrated that the pooled sensitivity of the six imaging techniques were sorted as follows: SWE > 18F-FCH PET/CT > 11C-choline PET/CT > TRTE > 11C-acetate PET/CT > 18F-FDG PET/CT; the pooled specificity were also compared: SWE > 18F-FCH PET/CT > 11C-choline PET/CT > TRTE > 18F-FDG PET/CT > 11C-acetate PET/CT; finally, the pooled diagnostic accuracy of the six imaging techniques based on AUC were ranked as below: SWE > 18F-FCH PET/CT > 11C-choline PET/CT > TRTE > 11C-acetate PET/CT > 18F-FDG PET/CT. SWE and 18F-FCH PET/CT imaging could offer more assistance in the early diagnosis of PCa than any other studied imaging techniques. However, the diagnostic ranking of the six imaging techniques might not be applicable to the clinical phase due to the shortage of stratified analysis.
Collapse
Affiliation(s)
- Qiaohong Ouyang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing, 100048, China.
| | - Zhongxiang Duan
- Department of Nuclear Medicine, The First Affiliated Hospital of Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing, 100048, China
| | - Jixiao Lei
- Department of Nuclear Medicine, The First Affiliated Hospital of Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing, 100048, China
| | - Guangli Jiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing, 100048, China
| |
Collapse
|
26
|
Pandit-Taskar N, O'Donoghue JA, Durack JC, Lyashchenko SK, Cheal SM, Beylergil V, Lefkowitz RA, Carrasquillo JA, Martinez DF, Fung AM, Solomon SB, Gönen M, Heller G, Loda M, Nanus DM, Tagawa ST, Feldman JL, Osborne JR, Lewis JS, Reuter VE, Weber WA, Bander NH, Scher HI, Larson SM, Morris MJ. A Phase I/II Study for Analytic Validation of 89Zr-J591 ImmunoPET as a Molecular Imaging Agent for Metastatic Prostate Cancer. Clin Cancer Res 2015; 21:5277-85. [PMID: 26175541 DOI: 10.1158/1078-0432.ccr-15-0552] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/28/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Standard imaging for assessing osseous metastases in advanced prostate cancer remains focused on altered bone metabolism and is inadequate for diagnostic, prognostic, or predictive purposes. We performed a first-in-human phase I/II study of (89)Zr-DFO-huJ591 ((89)Zr-J591) PET/CT immunoscintigraphy to assess performance characteristics for detecting metastases compared with conventional imaging modalities (CIM) and pathology. EXPERIMENTAL DESIGN Fifty patients with progressive metastatic castration-resistant prostate cancers were injected with 5 mCi of (89)Zr-J591. Whole-body PET/CT scans were obtained, and images were analyzed for tumor visualization. Comparison was made to contemporaneously obtained bone scintigraphy and cross-sectional imaging on a lesion-by-lesion basis and with biopsies of metastatic sites. RESULTS Median standardized uptake value for (89)Zr-J591-positive bone lesions (n = 491) was 8.9 and for soft-tissue lesions (n = 90), it was 4.8 (P < 0.00003). (89)Zr-J591 detected 491 osseous sites compared with 339 by MDP and 90 soft-tissue lesions compared with 124 by computed tomography (CT). Compared with all CIMs combined, (89)Zr-J591 detected an additional 99 osseous sites. Forty-six lesions (21 bone and 25 soft tissue) were biopsied in 34 patients; 18 of 19 (89)Zr-J591-positive osseous sites and 14 of 16 (89)Zr-J591-positive soft tissue sites were positive for prostate cancer. The overall accuracy of (89)Zr-J591 was 95.2% (20 of 21) for osseous lesions and 60% (15 of 25) for soft-tissue lesions. CONCLUSIONS (89)Zr-J591 imaging demonstrated superior targeting of bone lesions relative to CIMs. Targeting soft-tissue lesions was less optimal, although (89)Zr-J591 had similar accuracy as individual CIMs. This study will provide benchmark data for comparing performance of proposed prostate-specific membrane antigen (PSMA) targeting agents for prostate cancer.
Collapse
Affiliation(s)
- Neeta Pandit-Taskar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York.
| | - Joseph A O'Donoghue
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeremy C Durack
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Serge K Lyashchenko
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York. Radiochemistry and Molecular Imaging Probe Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah M Cheal
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Volkan Beylergil
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert A Lefkowitz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jorge A Carrasquillo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Danny F Martinez
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alex Mak Fung
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stephen B Solomon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Mithat Gönen
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Glenn Heller
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Massimo Loda
- Department of Pathology, Dana-Farber Cancer Institute; Brigham & Women's Hospital; and Broad Institute, Boston, Massachusetts
| | - David M Nanus
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Scott T Tagawa
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Jarett L Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph R Osborne
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Jason S Lewis
- Department of Radiology, Weill Cornell Medical College, New York, New York. Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York. Radiochemistry and Molecular Imaging Probe Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Neil H Bander
- Department of Medicine, Weill Cornell Medical College, New York, New York. Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Howard I Scher
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiology, Weill Cornell Medical College, New York, New York. Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
27
|
Popovics P, Frigo DE, Schally AV, Rick FG. Targeting the 5'-AMP-activated protein kinase and related metabolic pathways for the treatment of prostate cancer. Expert Opin Ther Targets 2015; 19:617-32. [PMID: 25600663 DOI: 10.1517/14728222.2015.1005603] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Increasing evidence suggests that prostate cancer cells undergo unique metabolic reprogramming during transformation. A master regulator of cellular homeostasis, 5'-AMP-activated protein kinase (AMPK), directs metabolic adaptation that supports the growth demands of rapidly dividing cancer cells. The utilization of AMPK as a therapeutic target may therefore provide an effective strategy in the treatment of prostate cancer. AREAS COVERED Our review describes the regulation of AMPK by androgens and upstream kinases including the calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) in prostate cancer. Oncogenic, AMPK-regulated pathways that direct various metabolic processes are also addressed. Furthermore, we discuss the role of AMPK in growth arrest and autophagy as a potential survival pathway for cancer cells. In addition, by regulating non-metabolic pathways, AMPK may stimulate migration and mitosis. Finally, this review summarizes efforts to treat prostate cancer with pharmacological agents capable of modulating AMPK signaling. EXPERT OPINION Current research is primarily focused on developing drugs that activate AMPK as a treatment for prostate cancer. However, oncogenic aspects of AMPK signaling calls for caution about employing such therapies. We think that inhibitors of CaMKK2 or AMPK, or perhaps the modulation of downstream targets of AMPK, will gain importance in the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Petra Popovics
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education , Research (151) 2A127, 1201 NW 16th St, Miami, FL 33125 , USA +1 305 5753477 ; +1 305 5753126 ;
| | | | | | | |
Collapse
|
28
|
[18F]Fluoromethylcholine as a Chemotherapy Response Read-Out in Prostate Cancer Cells. Mol Imaging Biol 2014; 17:319-27. [DOI: 10.1007/s11307-014-0803-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
29
|
Abstract
Kidney, bladder, and prostate cancer account for more than one-eighth of new cancer cases worldwide. Imaging in kidney cancer is dominated by computed tomography (CT). Positron emission tomography (PET) imaging of bladder cancer is hampered by the urinary excretion of the most common PET tracer, 18F-fluoro-deoxy-glucose (FDG). PET imaging has been applied more often in prostate cancer. FDG-PET/CT is claimed to have a high frequency of false-negative results in urologic cancers; however, this finding may instead reflect correctly the state of disease being due to slow-growing cancers with a good prognosis and without a need of therapy.
Collapse
|
30
|
Simoncic U, Perlman S, Liu G, Staab MJ, Straus JE, Jeraj R. Comparison of NaF and FDG PET/CT for assessment of treatment response in castration-resistant prostate cancers with osseous metastases. Clin Genitourin Cancer 2014; 13:e7-e17. [PMID: 25128349 DOI: 10.1016/j.clgc.2014.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 07/04/2014] [Accepted: 07/08/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Assessment of skeletal metastases' response to therapy is a highly relevant but unresolved clinical problem. The main goal of this work was to compare pharmacodynamic responses to therapy assessed with positron emission tomography-computed tomography (PET/CT) using fluorine-18 sodium fluoride (NaF) and fluorine-18 fluorodeoxyglucose (FDG) as the tracers. MATERIALS AND METHODS Patients with prostate cancer with known osseous metastases were treated with zibotentan (ZD4054) and imaged with combined dynamic NaF/FDG PET/CT before therapy (baseline), after 4 weeks of therapy (week 4), and after 2 weeks of treatment break (week 6). Kinetic analysis allowed comparison of the voxel-based tracer uptake rate parameter Ki, the vasculature parameters K1 (measuring perfusion/permeability) and Vb (measuring vasculature fraction in the tissue), and the standardized uptake values (SUVs). RESULTS Correlations were high for the NaF and FDG peak uptake parameters (Ki and SUV correlations ranged from 0.57 to 0.88) and for vasculature parameters (K1 and Vb correlations ranged from 0.61 to 0.81). Correlation was low between the NaF and FDG week 4 Ki responses (ρ = 0.35; P = .084) but was higher for NaF and FDG week 6 Ki responses (ρ = 0.72; P < .0001). Correlations for vasculature responses were always low (ρ < 0.35). NaF and FDG uptakes in the osseous metastases were spatially dislocated, with overlap in the range from 0% to 80%. CONCLUSION This study found that late NaF and FDG uptake responses are consistently correlated but that earlier uptake responses and all vasculature responses can be unrelated. This study also confirmed that FDG and NaF uptakes are spatially dislocated. Although treatment responses assessed with NaF and FDG may be correlated, using both tracers provides additional information.
Collapse
Affiliation(s)
- Urban Simoncic
- Jozef Stefan Institute, Ljubljana, Slovenia; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI; Centre of Excellence for Biosensors, Instrumentation and Process Control (COBIK), Ajdovscina, Slovenia.
| | - Scott Perlman
- Department of Radiology, University of Wisconsin-Madison, Madison, WI; University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Glenn Liu
- Genitourinary Oncology Research Program, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI; University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Mary Jane Staab
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Jane Elizabeth Straus
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Robert Jeraj
- Jozef Stefan Institute, Ljubljana, Slovenia; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI; Department of Radiology, University of Wisconsin-Madison, Madison, WI; Centre of Excellence for Biosensors, Instrumentation and Process Control (COBIK), Ajdovscina, Slovenia; University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| |
Collapse
|
31
|
Springer CS, Li X, Tudorica LA, Oh KY, Roy N, Chui SYC, Naik AM, Holtorf ML, Afzal A, Rooney WD, Huang W. Intratumor mapping of intracellular water lifetime: metabolic images of breast cancer? NMR IN BIOMEDICINE 2014; 27:760-73. [PMID: 24798066 PMCID: PMC4174415 DOI: 10.1002/nbm.3111] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 05/10/2023]
Abstract
Shutter-speed pharmacokinetic analysis of dynamic-contrast-enhanced (DCE)-MRI data allows evaluation of equilibrium inter-compartmental water interchange kinetics. The process measured here - transcytolemmal water exchange - is characterized by the mean intracellular water molecule lifetime (τi). The τi biomarker is a true intensive property not accessible by any formulation of the tracer pharmacokinetic paradigm, which inherently assumes it is effectively zero when applied to DCE-MRI. We present population-averaged in vivo human breast whole tumor τi changes induced by therapy, along with those of other pharmacokinetic parameters. In responding patients, the DCE parameters change significantly after only one neoadjuvant chemotherapy cycle: while K(trans) (measuring mostly contrast agent (CA) extravasation) and kep (CA intravasation rate constant) decrease, τi increases. However, high-resolution, (1 mm)(2), parametric maps exhibit significant intratumor heterogeneity, which is lost by averaging. A typical 400 ms τi value means a trans-membrane water cycling flux of 10(13) H2O molecules s(-1)/cell for a 12 µm diameter cell. Analyses of intratumor variations (and therapy-induced changes) of τi in combination with concomitant changes of ve (extracellular volume fraction) indicate that the former are dominated by alterations of the equilibrium cell membrane water permeability coefficient, PW, not of cell size. These can be interpreted in light of literature results showing that τi changes are dominated by a PW (active) component that reciprocally reflects the membrane driving P-type ATPase ion pump turnover. For mammalian cells, this is the Na(+), K(+)-ATPase pump. These results promise the potential to discriminate metabolic and microenvironmental states of regions within tumors in vivo, and their changes with therapy.
Collapse
Affiliation(s)
- Charles S Springer
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortland, OR, USA
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- *Correspondence to: C. S. Springer, Jr, Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, USA. E-mail:
| | - Xin Li
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortland, OR, USA
| | - Luminita A Tudorica
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- Department of Diagnostic Radiology, Oregon Health and Science UniversityPortland, OR, USA
| | - Karen Y Oh
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- Department of Diagnostic Radiology, Oregon Health and Science UniversityPortland, OR, USA
| | - Nicole Roy
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- Department of Diagnostic Radiology, Oregon Health and Science UniversityPortland, OR, USA
| | - Stephen Y-C Chui
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- Department of Hematology/Oncology, Oregon Health and Science UniversityPortland, OR, USA
| | - Arpana M Naik
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- Department of Surgical Oncology, Oregon Health and Science UniversityPortland, OR, USA
| | - Megan L Holtorf
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
- Clinical Trials Office, Oregon Health and Science UniversityPortland, OR, USA
| | - Aneela Afzal
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortland, OR, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortland, OR, USA
| | - Wei Huang
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortland, OR, USA
- Knight Cancer Institute, Oregon Health and Science UniversityPortland, OR, USA
| |
Collapse
|
32
|
Abstract
Imaging plays a central role in the detection, diagnosis, staging, and follow-up of prostate carcinoma. This article discusses the role of multiple imaging modalities in the diagnosis and staging of prostate cancer, with attention to imaging features of localized and metastatic disease, imaging adjuncts to improve prostate biopsy, and potential imaging biomarkers. In addition, the role of imaging in the management of prostate cancer, with emphasis on surveillance, evaluation of response to new therapies, and detection of recurrent disease is described. Lastly, future directions in prostate cancer imaging are presented.
Collapse
|
33
|
O'Sullivan F, Muzi M, Mankoff DA, Eary JF, Spence AM, Krohn KA. VOXEL-LEVEL MAPPING OF TRACER KINETICS IN PET STUDIES: A STATISTICAL APPROACH EMPHASIZING TISSUE LIFE TABLES. Ann Appl Stat 2014; 8:1065-1094. [PMID: 25392718 PMCID: PMC4225726 DOI: 10.1214/14-aoas732] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Most radiotracers used in dynamic positron emission tomography (PET) scanning act in a linear time-invariant fashion so that the measured time-course data are a convolution between the time course of the tracer in the arterial supply and the local tissue impulse response, known as the tissue residue function. In statistical terms the residue is a life table for the transit time of injected radiotracer atoms. The residue provides a description of the tracer kinetic information measurable by a dynamic PET scan. Decomposition of the residue function allows separation of rapid vascular kinetics from slower blood-tissue exchanges and tissue retention. For voxel-level analysis, we propose that residues be modeled by mixtures of nonparametrically derived basis residues obtained by segmentation of the full data volume. Spatial and temporal aspects of diagnostics associated with voxel-level model fitting are emphasized. Illustrative examples, some involving cancer imaging studies, are presented. Data from cerebral PET scanning with 18F fluoro-deoxyglucose (FDG) and 15O water (H2O) in normal subjects is used to evaluate the approach. Cross-validation is used to make regional comparisons between residues estimated using adaptive mixture models with more conventional compartmental modeling techniques. Simulations studies are used to theoretically examine mean square error performance and to explore the benefit of voxel-level analysis when the primary interest is a statistical summary of regional kinetics. The work highlights the contribution that multivariate analysis tools and life-table concepts can make in the recovery of local metabolic information from dynamic PET studies, particularly ones in which the assumptions of compartmental-like models, with residues that are sums of exponentials, might not be certain.
Collapse
|
34
|
Abstract
The availability of new therapeutic options for the treatment of metastatic castration-resistant prostate cancer (mCRPC) has heightened the importance of monitoring and assessing treatment response. Accordingly, there is an unmet clinical need for reliable biomarkers that can be used to guide therapy. Circulating tumour cells (CTCs) are rare cells that are shed from primary and metastatic tumour deposits into the peripheral circulation, and represent a means of performing noninvasive tumour sampling. Indeed, enumeration of CTCs before and after therapy has shown that CTC burden correlates with prognosis in patients with mCRPC. Moreover, studies have demonstrated the potential of molecular analysis of CTCs in monitoring and predicting response to therapy in patients. This Review describes the challenges associated with monitoring treatment response in mCRPC, and the advancements in CTC-analysis technologies applied to such assessments and, ultimately, guiding prostate cancer treatment.
Collapse
|
35
|
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]
|
36
|
Abstract
BACKGROUND Imaging of prostate carcinoma is an important adjunct to clinical evaluation and prostate specific antigen measurement for detecting metastases and tumor recurrence. In the past, the ability to assess intraprostatic tumor was limited. METHODS Pertinent literature was reviewed to describe the capabilities and limitations of the currently available imaging techniques for assessing prostate carcinoma. Evaluation of primary tumor and metastatic disease by ultrasonography, computed tomography (CT), magnetic resonance imaging (MRI), and nuclear medicine techniques is discussed. RESULTS Ultrasonography and MRI have limited usefulness for local staging of prostate cancer because of suboptimal sensitivity and specificity for identifying tumor extent and capsular penetration. Additional MRI techniques such as magnetic resonance-based perfusion imaging, diffusion imaging, and spectroscopy may provide incremental benefit. CT and bone scanning provide an assessment of metastatic disease but are also limited by the poor sensitivity of lymph node size as a criterion for detecting metastases. Novel imaging techniques such as hybrid imaging devices in the form of single-photon emission CT/CT gamma cameras, positron emission tomography/CT cameras, and, in the near future, positron emission tomography/MRI combined with tumor specific imaging radiotracers may have a significant impact on tumor staging and treatment response. CONCLUSIONS Cross-sectional imaging and scintigraphy have an important role in assessing prostate carcinoma metastases and treatment response. Increasingly, the incremental value of primary tumor imaging through MRI is being realized.
Collapse
Affiliation(s)
- Eric K Outwater
- Department of Diagnostic Imaging, Moffitt Cancer Center, Tampa, FL 33612, USA.
| | | |
Collapse
|
37
|
Mohsen B, Giorgio T, Rasoul ZS, Werner L, Ali GRM, Reza DKV, Ramin S. Application of C-11-acetate positron-emission tomography (PET) imaging in prostate cancer: systematic review and meta-analysis of the literature. BJU Int 2013; 112:1062-1072. [PMID: 23937453 DOI: 10.1111/bju.12279] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
To review the literature on the application of (11) C-acetate positron-emission tomography (PET) imaging in prostate cancer. We systematically reviewed the available literature and presented the results in meta-analysis format. PubMed, SCOPUS, ISI web of knowledge, Science Direct, Springer, and Google Scholar were searched with 'Acetate AND PET AND Prostate' as keywords. All studies that evaluated accuracy of (11) C-acetate imaging in primary or recurrent prostate cancer were included, if enough data could be extracted for calculation of sensitivity and/or specificity. In all, 23 studies were included in the study. For evaluation of primary tumour, pooled sensitivity was 75.1 (69.8-79.8)% and specificity was 75.8 (72.4-78.9)%. For detection of recurrence, sensitivity was 64 (59-69)% and specificity was 93 (83-98)%. Sensitivity for recurrence detection was higher in post-surgical vs post-radiotherapy patients and in patients with PSA at relapse of >1 ng/mL. Studies using PET/computed tomography vs PET also showed higher sensitivity for detection of recurrence. Imaging with (11) C-acetate PET can be useful in patients with prostate cancer. This is especially true for evaluation of patients at PSA relapse, although the sensitivity is overall low. For primary tumour evaluation (localisation of tumour in the prostate and differentiation of malignant from benign lesions), (11) C-acetate is of limited value due to low sensitivity and specificity. Due to the poor quality of the included studies, the results should be interpreted with caution and further high-quality studies are needed.
Collapse
Affiliation(s)
- Beheshti Mohsen
- PET-CT Center LINZ, St. Vincent's Hospital-Nuclear Medicine and Endocrinology, Linz, Austria
| | | | | | | | | | | | | |
Collapse
|
38
|
Tennakoon JB, Shi Y, Han JJ, Tsouko E, White MA, Burns AR, Zhang A, Xia X, Ilkayeva OR, Xin L, Ittmann MM, Rick FG, Schally AV, Frigo DE. Androgens regulate prostate cancer cell growth via an AMPK-PGC-1α-mediated metabolic switch. Oncogene 2013; 33:5251-61. [PMID: 24186207 PMCID: PMC4009392 DOI: 10.1038/onc.2013.463] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 08/28/2013] [Accepted: 09/30/2013] [Indexed: 12/25/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy among men in industrialized countries, accounting for the second leading cause of cancer-related deaths. Although we now know that the androgen receptor (AR) is important for progression to the deadly advanced stages of the disease, it is poorly understood what AR-regulated processes drive this pathology. Here we demonstrate that AR regulates prostate cancer cell growth via the metabolic sensor 5'-AMP-activated protein kinase (AMPK), a kinase that classically regulates cellular energy homeostasis. In patients, activation of AMPK correlated with prostate cancer progression. Using a combination of radiolabeled assays and emerging metabolomic approaches, we also show that prostate cancer cells respond to androgen treatment by increasing not only rates of glycolysis, as is commonly seen in many cancers, but also glucose and fatty acid oxidation. Importantly, this effect was dependent on androgen-mediated AMPK activity. Our results further indicate that the AMPK-mediated metabolic changes increased intracellular ATP levels and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)-mediated mitochondrial biogenesis, affording distinct growth advantages to the prostate cancer cells. Correspondingly, we used outlier analysis to determine that PGC-1α is overexpressed in a subpopulation of clinical cancer samples. This was in contrast to what was observed in immortalized benign human prostate cells and a testosterone-induced rat model of benign prostatic hyperplasia. Taken together, our findings converge to demonstrate that androgens can co-opt the AMPK-PGC-1α signaling cascade, a known homeostatic mechanism, to increase prostate cancer cell growth. The current study points to the potential utility of developing metabolic-targeted therapies directed toward the AMPK-PGC-1α signaling axis for the treatment of prostate cancer.
Collapse
Affiliation(s)
- J B Tennakoon
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Y Shi
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - J J Han
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - E Tsouko
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - M A White
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - A R Burns
- College of Optometry, University of Houston, Houston, TX, USA
| | - A Zhang
- Center for Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - X Xia
- Center for Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | - O R Ilkayeva
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - L Xin
- 1] Departments of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA [2] Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA [3] Dan L. Duncan Cancer Center, Houston, TX, USA
| | - M M Ittmann
- 1] Departments of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA [2] Dan L. Duncan Cancer Center, Houston, TX, USA [3] Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - F G Rick
- 1] Veterans Affairs Medical Center and South Florida VA Foundation for Research and Education, Miami, FL, USA [2] Department of Urology, Florida International University, Herbert Wertheim College of Medicine, Miami, FL, USA
| | - A V Schally
- 1] Veterans Affairs Medical Center and South Florida VA Foundation for Research and Education, Miami, FL, USA [2] Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, USA [3] Divisions of Hematology/Oncology, University of Miami, Miller School of Medicine, Miami, FL, USA [4] Division of Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - D E Frigo
- 1] Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA [2] Center for Genomic Medicine, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
39
|
Jacobson O, Chen X. Interrogating tumor metabolism and tumor microenvironments using molecular positron emission tomography imaging. Theranostic approaches to improve therapeutics. Pharmacol Rev 2013; 65:1214-56. [PMID: 24064460 PMCID: PMC3799232 DOI: 10.1124/pr.113.007625] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technology that is becoming increasingly important for the measurement of physiologic, biochemical, and pharmacological functions at cellular and molecular levels in patients with cancer. Formation, development, and aggressiveness of tumor involve a number of molecular pathways, including intrinsic tumor cell mutations and extrinsic interaction between tumor cells and the microenvironment. Currently, evaluation of these processes is mainly through biopsy, which is invasive and limited to the site of biopsy. Ongoing research on specific target molecules of the tumor and its microenvironment for PET imaging is showing great potential. To date, the use of PET for diagnosing local recurrence and metastatic sites of various cancers and evaluation of treatment response is mainly based on [(18)F]fluorodeoxyglucose ([(18)F]FDG), which measures glucose metabolism. However, [(18)F]FDG is not a target-specific PET tracer and does not give enough insight into tumor biology and/or its vulnerability to potential treatments. Hence, there is an increasing need for the development of selective biologic radiotracers that will yield specific biochemical information and allow for noninvasive molecular imaging. The possibility of cancer-associated targets for imaging will provide the opportunity to use PET for diagnosis and therapy response monitoring (theranostics) and thus personalized medicine. This article will focus on the review of non-[(18)F]FDG PET tracers for specific tumor biology processes and their preclinical and clinical applications.
Collapse
Affiliation(s)
- Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD.
| | | |
Collapse
|
40
|
Imaging of castration-resistant prostrate cancer: development of imaging response biomarkers. Curr Opin Urol 2013; 23:230-6. [PMID: 23422587 DOI: 10.1097/mou.0b013e32835e9edc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The current standard for imaging castration-resistant prostate cancer (CRPC) focuses solely on detection. However, in order to assess treatment response, imaging must provide quantitative results that can be validated. RECENT FINDINGS Bone scintigraphy remains the most commonly used imaging tool for CRPC in bone, but with limited quantification capabilities. Both PET and MRI provide quantitative measures that could be used to assess treatment response. Several PET tracers have been shown to be able to detect bone metastases, but more research regarding their use for treatment response assessment is necessary. Similarly, research has shown that diffusion-weighted and dynamic contrast-enhanced MRI can detect metastases, with some studies suggesting that they may be suitable for assessing treatment response. SUMMARY Recent research has shown that many imaging techniques are able to successfully detect metastases in CRPC patients as well as or better than standard imaging. These imaging methods can also be applied to treatment response assessment; however, more research must be done to validate the quantitative measures before these techniques can be used clinically for assessing patients.
Collapse
|
41
|
Kitajima K, Murphy RC, Nathan MA, Sugimura K. Update on positron emission tomography for imaging of prostate cancer. Int J Urol 2013; 21:12-23. [PMID: 23991644 DOI: 10.1111/iju.12250] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 07/10/2013] [Indexed: 12/11/2022]
Abstract
Prostate cancer is the most common non-cutaneous malignancy among men in the Western world, and continues to be a major health problem. Imaging has recently become more important in the clinical management of prostate cancer patients, including diagnosis, staging, choice of optimal treatment strategy, treatment follow up and restaging. Positron emission tomography, a functional and molecular imaging technique, has opened a new field in clinical oncological imaging. The most common positron emission tomography radiotracer, 18F-fluorodeoxyglucose, has been limited in imaging of prostate cancer. Recently, however, other positron emission tomography tracers, such as 11C-acetate and 11C- or (18) F-choline, have shown promising results. In the present review article, we overview the potential and current use of positron emission tomography or positron emission tomography/computed tomography imaging employing the four most commonly used positron emission tomography radiotracers, 18F-fluorodeoxyglucose, 11C-acetate and 11C- or 18F-choline, for imaging evaluation of prostate cancer.
Collapse
Affiliation(s)
- Kazuhiro Kitajima
- Department of Radiology, Kobe University School of Medicine, Kobe, Japan
| | | | | | | |
Collapse
|
42
|
Sharma P, Karunanithi S, Singh Dhull V, Jain S, Bal C, Kumar R. Prostate cancer with lytic bone metastases: 18F-fluorodeoxyglucose positron emission tomography-computed tomography for diagnosis and monitoring response to medical castration therapy. Indian J Nucl Med 2013; 28:178-179. [PMID: 24250030 PMCID: PMC3822421 DOI: 10.4103/0972-3919.119545] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Lytic bone metastases are rare in prostate cancer. We here present 18 fluorine fluorodeoxyglucose ((18)F-FDG) positron emission tomography computed tomography (PET-CT) images of a 67-year-old male patient with lytic metastases from prostate cancer. Repeat (18)F-FDG PET-CT done 6 months later showed response to medical castration therapy. While the role of (18)F-FDG PET-CT for sclerotic bone metastases in prostate cancer remains controversial, it appears to be useful for detection and response assessment of lytic prostate cancer metastases.
Collapse
Affiliation(s)
- Punit Sharma
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sellam Karunanithi
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Varun Singh Dhull
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sachin Jain
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
43
|
Sartor O, Eisenberger M, Kattan MW, Tombal B, Lecouvet F. Unmet needs in the prediction and detection of metastases in prostate cancer. Oncologist 2013; 18:549-57. [PMID: 23650019 DOI: 10.1634/theoncologist.2013-0027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The therapeutic landscape for the treatment of advanced prostate cancer is rapidly evolving, especially for those patients with metastatic castration-resistant prostate cancer (CPRC). Despite advances in therapy options, the diagnostic landscape has remained relatively static, with few guidelines or reviews addressing the optimal timing or methodology for the radiographic detection of metastatic disease. Given recent reports indicating a substantial proportion of patients with CRPC thought to be nonmetastatic (M0) are in fact metastatic (M1), there is now a clear opportunity and need for improvement in detection practices. Herein, we discuss the current status of predicting the presence of metastatic disease, with a particular emphasis on the detection of the M0 to M1 transition. In addition, we review current data on newer imaging technologies that are changing the way metastases are detected. Whether earlier detection of metastatic disease will ultimately improve patient outcomes is unknown, but given that the therapeutic options for those with metastatic and nonmetastatic CPRC vary, there are considerable implications of how and when metastases are detected.
Collapse
Affiliation(s)
- Oliver Sartor
- Tulane Cancer Center, New Orleans, Louisiana 70112, USA.
| | | | | | | | | |
Collapse
|
44
|
Sörensen J, Owenius R, Lax M, Johansson S. Regional distribution and kinetics of [18F]fluciclovine (anti-[18F]FACBC), a tracer of amino acid transport, in subjects with primary prostate cancer. Eur J Nucl Med Mol Imaging 2012. [PMID: 23208700 DOI: 10.1007/s00259-012-2291-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jens Sörensen
- Section of Nuclear Medicine and PET, Department of Radiology, Oncology and Radiation Sciences, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
45
|
Abstract
Functional radionuclide imaging modalities, now commonly combined with anatomical imaging modalities computed tomography (CT) or magnetic resonance imaging (single photon emission computed tomography [SPECT]/CT, positron emission tomography [PET]/CT, and PET/magnetic resonance imaging), are promising tools for the management of prostate cancer, particularly for therapeutic implications. Sensitive detection capability of prostate cancer using these imaging modalities is one issue; however, the treatment of prostate cancer using the information that can be obtained from functional radionuclide imaging techniques is another challenging area. There are not many SPECT or PET radiotracers that can cover the full spectrum of the management of prostate cancer from initial detection to staging, prognosis predictor, and all the way to treatment response assessment. However, when used appropriately, the information from functional radionuclide imaging improves, and sometimes significantly changes, the whole course of the cancer management. The limitations of using SPECT and PET radiotracers with regard to therapeutic implications are not so much different from their limitations solely for the task of detecting prostate cancer; however, the specific imaging target and how this target is reliably imaged by SPECT and PET can potentially make significant impact in the treatment of prostate cancer. Finally, although the localized prostate cancer is considered manageable, there is still significant need for improvement in noninvasive imaging of metastatic prostate cancer, in treatment guidance, and in response assessment from functional imaging, including radionuclide-based techniques. In this review article, we present the rationale of using functional radionuclide imaging and the therapeutic implications for each of radionuclide imaging agent that have been studied in human subjects.
Collapse
Affiliation(s)
- Carina Mari Aparici
- Department of Radiology and Biomedical Imaging, University of California, and Nuclear Medicine Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94107, USA
| | | |
Collapse
|
46
|
Abstract
OBJECTIVE Recent advances in the fundamental understanding of the complex biology of prostate cancer have provided an increasing number of potential targets for imaging and treatment. The imaging evaluation of prostate cancer needs to be tailored to the various phases of this remarkably heterogeneous disease. CONCLUSION In this article, I review the current state of affairs on a range of PET radiotracers for potential use in the imaging evaluation of men with prostate cancer.
Collapse
|
47
|
Abstract
The classification of clinical disease states within advanced prostate cancer is set apart from other solid tumors largely through measurement of prostate-specific antigen in the blood. This testing has allowed the distinction between the castration-sensitive and the castration-resistant states, to complement radiographic distinction within advanced prostate cancer. This has paved the way for advances in prognostication and treatment of patients within a heterogeneous disease group. Currently used clinical classifications have limitations and continue to evolve. The authors define the current disease states and discuss implications for prognosis and treatment decisions, as well as the limitations of existing classifications and emerging discoveries.
Collapse
Affiliation(s)
- Heather H Cheng
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA 98109, USA.
| | | | | |
Collapse
|
48
|
Kurland BF, Gerstner ER, Mountz JM, Schwartz LH, Ryan CW, Graham MM, Buatti JM, Fennessy FM, Eikman EA, Kumar V, Forster KM, Wahl RL, Lieberman FS. Promise and pitfalls of quantitative imaging in oncology clinical trials. Magn Reson Imaging 2012; 30:1301-12. [PMID: 22898682 DOI: 10.1016/j.mri.2012.06.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 06/07/2012] [Accepted: 06/16/2012] [Indexed: 01/13/2023]
Abstract
Quantitative imaging using computed tomography, magnetic resonance imaging and positron emission tomography modalities will play an increasingly important role in the design of oncology trials addressing molecularly targeted, personalized therapies. The advent of molecularly targeted therapies, exemplified by antiangiogenic drugs, creates new complexities in the assessment of response. The Quantitative Imaging Network addresses the need for imaging modalities which can accurately and reproducibly measure not just change in tumor size but changes in relevant metabolic parameters, modulation of relevant signaling pathways, drug delivery to tumor and differentiation of apoptotic cell death from other changes in tumor volume. This article provides an overview of the applications of quantitative imaging to phase 0 through phase 3 oncology trials. We describe the use of a range of quantitative imaging modalities in specific tumor types including malignant gliomas, lung cancer, head and neck cancer, lymphoma, breast cancer, prostate cancer and sarcoma. In the concluding section, we discuss potential constraints on clinical trials using quantitative imaging, including complexity of trial conduct, impact on subject recruitment, incremental costs and institutional barriers. Strategies for overcoming these constraints are presented.
Collapse
|
49
|
Valotassiou V, Leondi A, Angelidis G, Psimadas D, Georgoulias P. SPECT and PET imaging of meningiomas. ScientificWorldJournal 2012; 2012:412580. [PMID: 22623896 PMCID: PMC3353476 DOI: 10.1100/2012/412580] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 01/26/2012] [Indexed: 12/21/2022] Open
Abstract
Meningiomas arise from the meningothelial cells of the arachnoid membranes. They are the most common primary intracranial neoplasms and represent about 20% of all intracranial tumors. They are usually diagnosed after the third decade of life and they are more frequent in women than in men. According to the World Health Organization (WHO) criteria, meningiomas can be classified into grade I meningiomas, which are benign, grade II (atypical) and grade III (anaplastic) meningiomas, which have a much more aggressive clinical behaviour. Computed Tomography (CT) and Magnetic Resonance Imaging (MRI) are routinely used in the diagnostic workup of patients with meningiomas. Molecular Nuclear Medicine Imaging with Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET) could provide complementary information to CT and MRI. Various SPECT and PET tracers may provide information about cellular processes and biological characteristics of meningiomas. Therefore, SPECT and PET imaging could be used for the preoperative noninvasive diagnosis and differential diagnosis of meningiomas, prediction of tumor grade and tumor recurrence, response to treatment, target volume delineation for radiation therapy planning, and distinction between residual or recurrent tumour from scar tissue.
Collapse
Affiliation(s)
- Varvara Valotassiou
- Nuclear Medicine Department, University Hospital of Larissa, Mezourlo, Larissa, Greece.
| | | | | | | | | |
Collapse
|
50
|
Current World Literature. Curr Opin Support Palliat Care 2012; 6:109-25. [DOI: 10.1097/spc.0b013e328350f70c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|