1
|
Zirakchian Zadeh M. PET/CT in assessment of colorectal liver metastases: a comprehensive review with emphasis on 18F-FDG. Clin Exp Metastasis 2023; 40:465-491. [PMID: 37682423 DOI: 10.1007/s10585-023-10231-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023]
Abstract
Approximately 25% of those who are diagnosed with colorectal cancer will develop colorectal liver metastases (CRLM) as their illness advances. Despite major improvements in both diagnostic and treatment methods, the prognosis for patients with CRLM is still poor, with low survival rates. Accurate employment of imaging methods is critical in identifying the most effective treatment approach for CRLM. Different imaging modalities are used to evaluate CRLM, including positron emission tomography (PET)/computed tomography (CT). Among the PET radiotracers, fluoro-18-deoxyglucose (18F-FDG), a glucose analog, is commonly used as the primary radiotracer in assessment of CRLM. As the importance of 18F-FDG-PET/CT continues to grow in assessment of CRLM, developing a comprehensive understanding of this subject becomes imperative for healthcare professionals from diverse disciplines. The primary aim of this article is to offer a simplified and comprehensive explanation of PET/CT in the evaluation of CRLM, with a deliberate effort to minimize the use of technical nuclear medicine terminology. This approach intends to provide various healthcare professionals and researchers with a thorough understanding of the subject matter.
Collapse
Affiliation(s)
- Mahdi Zirakchian Zadeh
- Molecular Imaging and Therapy and Interventional Radiology Services, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
2
|
Nak D, Küçük NÖ, Çelebioğlu EC, Bilgiç MS, Hayme S, Kır KM. The Role of 18F-FLT PET/CT in Assessing Early Response to Transarterial Radioembolization and Chemoembolization in Patients with Primary and Metastatic Liver Tumors. Mol Imaging Radionucl Ther 2022; 31:207-215. [PMID: 36268887 DOI: 10.4274/mirt.galenos.2022.85579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objectives Metastases and primary malignancies are common in the liver. Local ablative applications such as transarterial chemoembolization (TACE), and transarterial radioembolization (TARE) provide minimally invasive and safe treatment in unresectable liver tumors. Early detection of response to treatment prevents unnecessary toxicity and cost in non-responder patients and provides an earlier use of other options that may be effective. This study aimed to identify the role of 18F-fluorothymidine (FLT) positron emission tomography/computed tomography (PET/CT) in the assessment of early response to TACE and TARE treatments in patients with unresectable primary and metastatic liver tumors. Methods This single-center study included 63 patients who underwent 18F-FLT PET/CT for response evaluation after TACE and TARE. After excluding 20 patients whose data were missing 43 TARE-receiving patients were analyzed. The compatibility of change in semi-quantitative values obtained from the 18F-FLT PET/CT images with the treatment responses detected in 18F-fluorodeoxyglucose PET/CT, CT, and MR images and survival was evaluated. Results There was no correlation between early metabolic, morphological response, and 18F-FLT uptake pattern, and change in standardized uptake values (SUV) which were ΔSUVmax, ΔSUVmean, ΔSUVpeak., ΔSUVmean, Δ SUVpeak values. There was no significant correlation between 18F-FLT uptake pattern, ΔSUVmax, ΔSUVmean, ΔSUVpeak, and overall survival, progression-free survival (PFS) for the target lobe PFS for the whole-body. The survival distributions for the patients with >30% change in Δ SUVmax and ΔSUVpeak values were statistically significantly longer than the patients with <30% change (p<0.009 and p<0.024, respectively). Conclusion There was significant longer PFS for target liver lobe in patients with more than 30% decrease in 18F-FLT SUVmax and SUVpeak of the liver lesion in primary and metastatic unresectable liver tumors undergoing TARE.
Collapse
Affiliation(s)
- Demet Nak
- Recep Tayyip Erdoğan Training and Research Hospital, Clinic of Nuclear Medicine, Rize, Turkey
| | - Nuriye Özlem Küçük
- Ankara University Faculty of Medicine, Department of Nuclear Medicine, Ankara, Turkey
| | - Emre Can Çelebioğlu
- Ankara University Faculty of Medicine, Department of Radiology, Ankara, Turkey
| | - Mehmet Sadık Bilgiç
- Ankara University Faculty of Medicine, Department of Radiology, Ankara, Turkey
| | - Serhat Hayme
- Erzincan Binali Yıldırım University, Department of Biostatistics and Medical Informatics, Erzincan, Turkey
| | - Kemal Metin Kır
- Ankara University Faculty of Medicine, Department of Nuclear Medicine, Ankara, Turkey
| |
Collapse
|
4
|
Sorace AG, Elkassem AA, Galgano SJ, Lapi SE, Larimer BM, Partridge SC, Quarles CC, Reeves K, Napier TS, Song PN, Yankeelov TE, Woodard S, Smith AD. Imaging for Response Assessment in Cancer Clinical Trials. Semin Nucl Med 2020; 50:488-504. [PMID: 33059819 PMCID: PMC7573201 DOI: 10.1053/j.semnuclmed.2020.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The use of biomarkers is integral to the routine management of cancer patients, including diagnosis of disease, clinical staging and response to therapeutic intervention. Advanced imaging metrics with computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET) are used to assess response during new drug development and in cancer research for predictive metrics of response. Key components and challenges to identifying an appropriate imaging biomarker are selection of integral vs integrated biomarkers, choosing an appropriate endpoint and modality, and standardization of the imaging biomarkers for cooperative and multicenter trials. Imaging biomarkers lean on the original proposed quantified metrics derived from imaging such as tumor size or longest dimension, with the most commonly implemented metrics in clinical trials coming from the Response Evaluation Criteria in Solid Tumors (RECIST) criteria, and then adapted versions such as immune-RECIST (iRECIST) and Positron Emission Tomography Response Criteria in Solid Tumors (PERCIST) for immunotherapy response and PET imaging, respectively. There have been many widely adopted biomarkers in clinical trials derived from MRI including metrics that describe cellularity and vascularity from diffusion-weighted (DW)-MRI apparent diffusion coefficient (ADC) and Dynamic Susceptibility Contrast (DSC) or dynamic contrast enhanced (DCE)-MRI (Ktrans, relative cerebral blood volume (rCBV)), respectively. Furthermore, Fluorodexoyglucose (FDG), fluorothymidine (FLT), and fluoromisonidazole (FMISO)-PET imaging, which describe molecular markers of glucose metabolism, proliferation and hypoxia have been implemented into various cancer types to assess therapeutic response to a wide variety of targeted- and chemotherapies. Recently, there have been many functional and molecular novel imaging biomarkers that are being developed that are rapidly being integrated into clinical trials (with anticipation of being implemented into clinical workflow in the future), such as artificial intelligence (AI) and machine learning computational strategies, antibody and peptide specific molecular imaging, and advanced diffusion MRI. These include prostate-specific membrane antigen (PSMA) and trastuzumab-PET, vascular tumor burden extracted from contrast-enhanced CT, diffusion kurtosis imaging, and CD8 or Granzyme B PET imaging. Further excitement surrounds theranostic procedures such as the combination of 68Ga/111In- and 177Lu-DOTATATE to use integral biomarkers to direct care and personalize therapy. However, there are many challenges in the implementation of imaging biomarkers that remains, including understand the accuracy, repeatability and reproducibility of both acquisition and analysis of these imaging biomarkers. Despite the challenges associated with the biological and technical validation of novel imaging biomarkers, a distinct roadmap has been created that is being implemented into many clinical trials to advance the development and implementation to create specific and sensitive novel imaging biomarkers of therapeutic response to continue to transform medical oncology.
Collapse
Affiliation(s)
- Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL.
| | - Asser A Elkassem
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | - Suzanne E Lapi
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL; Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL
| | - Benjamin M Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| | | | - C Chad Quarles
- Division of Neuroimaging Research, Barrow Neurological Institute, Phoenix, AZ
| | - Kirsten Reeves
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; Cancer Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Tiara S Napier
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; Cancer Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Patrick N Song
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX; Department of Diagnostic Medicine, University of Texas at Austin, Austin, TX; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX
| | - Stefanie Woodard
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL
| | - Andrew D Smith
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
5
|
Kim JE, Chae SY, Kim JH, Kim HJ, Kim TW, Kim KP, Kim SY, Lee JL, Oh SJ, Kim JS, Ryu JS, Moon DH, Hong YS. 3′-Deoxy-3’-18F-Fluorothymidine and 18F-Fluorodeoxyglucose positron emission tomography for the early prediction of response to Regorafenib in patients with metastatic colorectal cancer refractory to all standard therapies. Eur J Nucl Med Mol Imaging 2019; 46:1713-1722. [DOI: 10.1007/s00259-019-04330-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 04/02/2019] [Indexed: 01/07/2023]
|
6
|
Buder-Bakhaya K, Hassel JC. Biomarkers for Clinical Benefit of Immune Checkpoint Inhibitor Treatment-A Review From the Melanoma Perspective and Beyond. Front Immunol 2018; 9:1474. [PMID: 30002656 PMCID: PMC6031714 DOI: 10.3389/fimmu.2018.01474] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibition (ICI) with anti-CTLA-4 and/or anti-PD-1 antibodies is standard treatment for metastatic melanoma. Anti-PD-1 (pembrolizumab, nivolumab) and anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) have been approved for treatment of several other advanced malignancies, including non-small-cell lung cancer (NSCLC); renal cell, and urothelial carcinoma; head and neck cancer; gastric, hepatocellular, and Merkel-cell carcinoma; and classical Hodgkin lymphoma. In some of these malignancies approval was based on the detection of biomarkers such as PD-L1 expression or high microsatellite instability. Methods We review the current status of prognostic and predictive biomarkers used in ICI for melanoma and other malignancies. We include clinical, tissue, blood, and stool biomarkers, as well as imaging biomarkers. Results Several biomarkers have been studied in ICI for metastatic melanoma. In clinical practice, pre-treatment tumor burden measured by means of imaging and serum lactate dehydrogenase level is already being used to estimate the likelihood of effective ICI treatment. In peripheral blood, the number of different immune cell types, such as lymphocytes, neutrophils, and eosinophils, as well as different soluble factors, have been correlated with clinical outcome. For intra-tumoral biomarkers, expression of the PD-1 ligand PD-L1 has been found to be of some predictive value for anti-PD-1-directed therapy for NSCLC and melanoma. A high mutational load, particularly when accompanied by neoantigens, seems to facilitate immune response and correlates with patient survival for all entities treated by use of ICI. Tumor microenvironment also seems to be of major importance. Interestingly, even the gut microbiome has been found to correlate with response to ICI, most likely through immuno-stimulatory effects of distinct bacteria. New imaging biomarkers, e.g., for PET, and magnetic resonance imaging are also being investigated, and results suggest they will make early prediction of patient response possible. Conclusion Several promising results are available regarding possible biomarkers for response to ICI, which need to be validated in large clinical trials. A better understanding of how ICI works will enable the development of biomarkers that can predict the response of individual patients.
Collapse
Affiliation(s)
- Kristina Buder-Bakhaya
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|