1
|
Nanni C, Deroose CM, Balogova S, Lapa C, Withofs N, Subesinghe M, Jamet B, Zamagni E, Ippolito D, Delforge M, Kraeber-Bodéré F. EANM guidelines on the use of [ 18F]FDG PET/CT in diagnosis, staging, prognostication, therapy assessment, and restaging of plasma cell disorders. Eur J Nucl Med Mol Imaging 2024; 52:171-192. [PMID: 39207486 PMCID: PMC11599630 DOI: 10.1007/s00259-024-06858-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024]
Abstract
We provide updated guidance and standards for the indication, acquisition, and interpretation of [18F]FDG PET/CT for plasma cell disorders. Procedures and characteristics are reported and different scenarios for the clinical use of [18F]FDG PET/CT are discussed. This document provides clinicians and technicians with the best available evidence to support the implementation of [18F]FDG PET/CT imaging in routine practice and future research.
Collapse
Affiliation(s)
- Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals (UZ) Leuven, 3000, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sona Balogova
- Nuclear Medicine, Comenius University, Bratislava, Slovakia
- Médecine Nucléaire, Hôpital Tenon, GH AP.SU, Paris, France
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Nadia Withofs
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liege, Belgium
- GIGA-CRC in Vivo Imaging, University of Liege, Liege, Belgium
| | - Manil Subesinghe
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Bastien Jamet
- Médecine Nucléaire, CHU Nantes, F-44000, Nantes, France
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy.
| | - Davide Ippolito
- Department of Diagnostic Radiology, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi 33, 20900, Monza, Italy
- University of Milano-Bicocca, School of Medicine, Via Cadore 33, 20090, Monza, Italy
| | | | | |
Collapse
|
2
|
Zhou HX, Chen WM. [Minimal residual disease assessment and progress in multiple myeloma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2024; 45:203-208. [PMID: 38604801 PMCID: PMC11078683 DOI: 10.3760/cma.j.cn121090-20230728-00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Indexed: 04/13/2024]
Abstract
With the rapid iteration of multiple myeloma therapeutics over the last two decades, as well as increasing remission rates and depth of remission in patients, traditional methods for monitoring disease response are insufficient to meet the clinical needs of new drugs. Minimal residual disease (MRD) is a more sensitive test for determining the depth of response, and data from multiple clinical trials and meta-analyses show that a negative MRD correlates with a better prognosis than a traditional complete response. MM is at the forefront of MRD evaluation and treatment. MRD detection methods have been continuously updated. The current MRD assessment has three dimensions: bone marrow-based MRD testing, MRD testing based on images of residual metabolic of focal lesions, and peripheral blood-based MRD testing. The various MRD assessment methods complement one another. The goal of this article is to discuss the currently used MRD assays, the progress, and challenges of MRD in MM, and to provide a reference for clinicians to better use the techniques.
Collapse
Affiliation(s)
- H X Zhou
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Multiple Myeloma Research Center of Beijing, Beijing 100020, China
| | - W M Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Multiple Myeloma Research Center of Beijing, Beijing 100020, China
| |
Collapse
|
3
|
Zirakchian Zadeh M. The role of conventional and novel PET radiotracers in assessment of myeloma bone disease. Bone 2024; 179:116957. [PMID: 37972747 DOI: 10.1016/j.bone.2023.116957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Over 80 % of patients with multiple myeloma (MM) experience osteolytic bone lesions, primarily due to an imbalanced interaction between osteoclasts and osteoblasts. This imbalance can lead to several adverse outcomes such as pain, fractures, limited mobility, and neurological impairments. Myeloma bone disease (MBD) raises the expense of management in addition to being a major source of disability and morbidity in myeloma patients. Whole-body x-ray radiography was the gold standard imaging modality for detecting lytic lesions. Osteolytic lesions are difficult to identify at an earlier stage on X-ray since the lesions do not manifest themselves on conventional radiographs until at least 30 % to 50 % of the bone mass has been destroyed. Hence, early diagnosis of osteolytic lesions necessitates the utilization of more complex and advanced imaging modalities, such as PET. One of the PET radiotracers that has been frequently investigated in MM is 18F-FDG, which has demonstrated a high level of sensitivity and specificity in detecting myeloma lesions. However, 18F-FDG PET/CT has several restrictions, and therefore the novel PET tracers that can overcome the limitations of 18F-FDG PET/CT should be further examined in assessment of MBD. The objective of this review article is to thoroughly examine the significance of both conventional and novel PET radiotracers in the assessment of MBD. The intention is to present the information in a manner that would be easily understood by healthcare professionals from diverse backgrounds, while minimizing the use of complex nuclear medicine terminology.
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
|
4
|
Nørgaard JN, Abildgaard N, Lysén A, Tsykunova G, Vangsted AJ, João C, Remen N, Nielsen LK, Osnes L, Stokke C, Connelly JP, Revheim MER, Schjesvold F. Intensifying treatment in PET-positive multiple myeloma patients after upfront autologous stem cell transplantation. Leukemia 2023; 37:2107-2114. [PMID: 37568010 DOI: 10.1038/s41375-023-01998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/26/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
18F-Fluorodeoxyglucose positron emission tomography/computed tomography (PET) positivity after first-line treatment with autologous stem cell transplantation (ASCT) in multiple myeloma is strongly correlated with reduced progression-free and overall survival. However, PET-positive patients who achieve PET negativity after treatment seem to have comparable outcomes to patients who were PET negative at diagnosis. Hence, giving PET-positive patients additional treatment may improve their outcome. In this phase II study, we screened first-line patients with very good partial response (VGPR) or better after ASCT with PET. PET-positive patients received four 28-day cycles of carfilzomib-lenalidomide-dexamethasone (KRd). Flow cytometry-based minimal residual disease (MRD) analysis was performed before and after treatment for correlation with PET. Overall, 159 patients were screened with PET. A total of 53 patients (33%) were PET positive and 57% of PET-positive patients were MRD negative, demonstrating that these response assessments are complementary. KRd consolidation converted 33% of PET-positive patients into PET negativity. MRD-negative patients were more likely to convert than MRD-positive patients. In summary, PET after ASCT detected residual disease in a substantial proportion of patients in VGPR or better, even in patients who were MRD negative, and KRd consolidation treatment changed PET status in 33% of patients.
Collapse
Affiliation(s)
- Jakob Nordberg Nørgaard
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- KG Jebsen Center for B cell malignancies, University of Oslo, Oslo, Norway.
| | - Niels Abildgaard
- Department of Hematology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anna Lysén
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Center for B cell malignancies, University of Oslo, Oslo, Norway
| | - Galina Tsykunova
- Division of Hematology, Haukeland University Hospital, Bergen, Norway
| | | | - Cristina João
- Department of Hematology, Champalimaud Centre for the Unknown, Lisboa, Portugal
- Nova Medical School, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Nora Remen
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
| | - Lene Kongsgaard Nielsen
- Quality of Life Research Center, Department of Haematology, Odense University Hospital, Odense, Denmark
- Department of Hematology, Gødstrup Hospital, Herning, Denmark
- OPEN, Open Patient data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Liv Osnes
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Caroline Stokke
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - James P Connelly
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Mona-Elisabeth R Revheim
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- The Intervention Centre, Oslo University Hospital, Oslo, Norway
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Center for B cell malignancies, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Kraeber-Bodéré F, Jamet B, Bezzi D, Zamagni E, Moreau P, Nanni C. New Developments in Myeloma Treatment and Response Assessment. J Nucl Med 2023; 64:1331-1343. [PMID: 37591548 PMCID: PMC10478822 DOI: 10.2967/jnumed.122.264972] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/06/2023] [Indexed: 08/19/2023] Open
Abstract
Recent innovative strategies have dramatically redefined the therapeutic landscape for treating multiple myeloma patients. In particular, the development and application of immunotherapy and high-dose therapy have demonstrated high response rates and have prolonged remission duration. Over the past decade, new morphologic or hybrid imaging techniques have gradually replaced conventional skeletal surveys. PET/CT using 18F-FDG is a powerful imaging tool for the workup at diagnosis and for therapeutic evaluation allowing medullary and extramedullary assessment. The independent negative prognostic value for progression-free and overall survival derived from baseline PET-derived parameters such as the presence of extramedullary disease or paramedullary disease, as well as the number of focal bone lesions and SUVmax, has been reported in several large prospective studies. During therapeutic evaluation, 18F-FDG PET/CT is considered the reference imaging technique because it can be performed much earlier than MRI, which lacks specificity. Persistence of significant abnormal 18F-FDG uptake after therapy is an independent negative prognostic factor, and 18F-FDG PET/CT and medullary flow cytometry are complementary tools for detecting minimal residual disease before maintenance therapy. The definition of a PET metabolic complete response has recently been standardized and the interpretation criteria harmonized. The development of advanced PET analysis and radiomics using machine learning, as well as hybrid imaging with PET/MRI, offers new perspectives for multiple myeloma imaging. Most recently, innovative radiopharmaceuticals such as C-X-C chemokine receptor type 4-targeted small molecules and anti-CD38 radiolabeled antibodies have shown promising results for tumor phenotype imaging and as potential theranostics.
Collapse
Affiliation(s)
- Françoise Kraeber-Bodéré
- Médecine nucléaire, CHU Nantes, Nantes Université, Université Angers, INSERM, CNRS, CRCI2NA, F-44000, Nantes, France
| | - Bastien Jamet
- Médecine nucléaire, CHU Nantes, F-44000, Nantes, France
| | - Davide Bezzi
- Department of Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna. Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Philippe Moreau
- Hématologie, CHU Nantes, Nantes Université, Université Angers, INSERM, CNRS, CRCI2NA, F-44000, Nantes, France; and
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
6
|
Li J, Ni B, Yu X, Wang C, Li L, Zhou Y, Gu Y, Huang G, Hou J, Liu J, Chen Y. Metabolic kinetic modeling of [ 11C]methionine based on total-body PET in multiple myeloma. Eur J Nucl Med Mol Imaging 2023; 50:2683-2691. [PMID: 37039900 DOI: 10.1007/s00259-023-06219-y] [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: 08/09/2022] [Accepted: 04/02/2023] [Indexed: 04/12/2023]
Abstract
PURPOSE Multiple myeloma (MM) is a malignant disease characterized by the secretion of monoclonal immunoglobulins and has a high demand for amino acids. [11C]methionine total-body PET is capable of noninvasive dynamic monitoring of radiotracer in vivo, thus providing a way to reveal the dynamic changes of myeloma metabolism. This study aims to analyze the metabolic process of [11C]methionine based on kinetic modeling, and to preliminary reveal its application value in MM. METHODS Dynamic total-body [11C]methionine PET/CT was conducted with uEXPLORER in 12 subjects (9 MM patients and 3 controls). The tissue time activity curves (TACs) of organs and bone marrows were extracted. Model fitting of TACs was operated using PMOD Kinetic Modeling. After validation by Goodness of fit (GOF), the reversible two-tissue compartment model (2T4k) was used to further analysis. R software was used to analyze the correlation between kinetic parameters and clinical indicators. RESULTS The 2T4k has passed the criterion of GOF and was used to fit the data of 0-20 minutes. The [11C]methionine net uptake rate (Ki) was significantly higher in the MM lesions than in the non-myeloma controls (control: 0.040±0.007 mL/g/min, MM: 0.171±0.108 mL/g/min, p=0.009). The Ki values were found to be correlated with M protein levels in MM patients. MM patients with t(4;14) translocations had an elevated k4 value compared with t(4;14) negative patients. CONCLUSION MM lesions have a propensity for uptake of [11C]methionine. The serum levels of M protein are correlated with [11C]methionine uptake rate in myeloma. Metabolic classification based on the k4 value may be a promising strategy for risk stratification in MM.
Collapse
Affiliation(s)
- Jiajin Li
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Beiwen Ni
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaofeng Yu
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Cheng Wang
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lianghua Li
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, 200032, China
| | - Yue Gu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, 200032, China
| | - Gang Huang
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jianjun Liu
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Yumei Chen
- Department of Nuclear Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
7
|
Filippi L, Frantellizzi V, Bartoletti P, Vincentis GD, Schillaci O, Evangelista L. Head-to-Head Comparison between FDG and 11C-Methionine in Multiple Myeloma: A Systematic Review. Diagnostics (Basel) 2023; 13:2009. [PMID: 37370904 DOI: 10.3390/diagnostics13122009] [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: 04/15/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The aim of this systematic review is to provide a comprehensive overview of the existing literature, comparing 18F-fluorodeoxyglucose (FDG) and 11C-methionine (MET) for the imaging of multiple myeloma (MM) with positron emission computed tomography (PET/CT). Relevant studies published from 2013 up to March 2023 were selected by searching Scopus, PubMed, and Web of Science. Selected imaging studies were analyzed using a modified version of the critical Appraisal Skills Programme (CASP). Ten studies encompassing 335 patients were selected. On a patient-based analysis, MET sensitivity ranged between 75.6% and 100%, resulting higher than that measured for FDG (0-100%). MET outperformed FDG for the detection of focal lesions, diffuse bone marrow involvement and mixed patterns. PET-derived parameters resulted higher for MET than for FDG, with a strong correlation with clinical variables (e.g., monoclonal component and beta-2-microglobulin levels, bone marrow infiltration, etc.), although FDG maintained a prognostic impact on outcome prediction. When compared to other tracers or imaging modalities, MET showed stronger correlation and inter-observer agreement than FDG. Although biased by the small cohorts and requiring confirmation through multicenter studies, preliminary findings suggest that MET-PET should be preferred to FDG for PET imaging of MM, or alternatively used as a complementary imaging modality. Some issues, such as tracer availability and the role of MET with respect to other emerging tracers (i.e., 68Ga-pentixafor, 18F-FACBC and 18F-FET), should be the topic of further investigations.
Collapse
Affiliation(s)
- Luca Filippi
- Nuclear Medicine Unit, "Santa Maria Goretti" Hospital, Via Antonio Canova, 04100 Latina, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Paola Bartoletti
- Nuclear Medicine Unit, Department of Medicine (DIMED), University of Padua, Via Giustiniani, 35128 Padua, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Viale Oxford 81, 00133 Rome, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
8
|
Bezzi D, Ambrosini V, Nanni C. Clinical Value of FDG-PET/CT in Multiple Myeloma: An Update. Semin Nucl Med 2023; 53:352-370. [PMID: 36446644 DOI: 10.1053/j.semnuclmed.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/28/2022]
Abstract
FDG-PET/CT is a standardized imaging technique that has reached a great importance in the management of patients affected by Multiple Myeloma. It is proved, in fact, that it allows a deep evaluation of therapy efficacy and provides several prognostic indexes both at staging and after therapy. For this reason, it is now recognised as a gold standard for therapy assessment. Beside this, in reacent years FDG-PET/CT contribution to the understanding of Multiple Myeloma has progressively grown. Papers have been published analyzing the prognostic value of active disease volume measurement and standardization issues, the meaning of FDG positive paramedullary and extrameduallary disease, the prognostic impact of FDG positive minimal residual disease, the relation between focal lesions and clonal eterogenity of this disease and the comparison with whole body DWI-MR in terms of detection and therapy assessment. These newer aspects not of clinical impact yet, of FDG-PET/CT in Multiple Myeloma will be presented and discussed in this review.
Collapse
Affiliation(s)
- Davide Bezzi
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Valentina Ambrosini
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy; Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| |
Collapse
|
9
|
FDG-PET/CT and new radiopharmaceuticals in patients with multiple myeloma. Rev Esp Med Nucl Imagen Mol 2023; 42:129-135. [PMID: 36746240 DOI: 10.1016/j.remnie.2023.01.004] [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: 12/13/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023]
Abstract
Multiple myeloma is a monoclonal gammopathy, a clonal proliferative disorder of plasma cells that produces a protein called M or myeloma protein in the bone marrow, usually IgG or IgA. It accounts for 1% in the general cancer statistics and represents 10% of all hematologic tumours, with a cumulative incidence in Spain of about 5/100,000/year. The incidence increases with age, so that 50% of cases are diagnosed in patients over 75 years of age, being infrequent in the population under 40 years of age. This publication details the indications of FDG PET/CT for the staging and response assessment in patients with MM, accepted by the international working group on myeloma, as well as new molecular imaging radiopharmaceuticals with potential value for personalised medicine.
Collapse
|
10
|
PET/TC con FDG y nuevos radiofármacos en pacientes con mieloma múltiple. Rev Esp Med Nucl Imagen Mol 2023. [DOI: 10.1016/j.remn.2023.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
|
11
|
Wang M, Zhao A, Li M, Niu T. Amino acids in hematologic malignancies: Current status and future perspective. Front Nutr 2023; 10:1113228. [PMID: 37032776 PMCID: PMC10076797 DOI: 10.3389/fnut.2023.1113228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
In recent years, growing emphasis has been placed on amino acids and their role in hematologic malignancies. Cancer cell metabolism is altered during tumorigenesis and development to meet expanding energetic and biosynthetic demands. Amino acids not only act as energy-supplying substances, but also play a vital role via regulating key signaling pathways, modulating epigenetic factors and remodeling tumor microenvironment. Targeting amino acids may be an effective therapeutic approach to address the current therapeutic challenges. Here, we provide an updated overview of mechanisms by which amino acids facilitate tumor development and therapy resistance. We also summarize novel therapies targeting amino acids, focusing on recent advances in basic research and their potential clinical implications.
Collapse
|
12
|
Islam A, Shaukat Z, Hussain R, Gregory SL. One-Carbon and Polyamine Metabolism as Cancer Therapy Targets. Biomolecules 2022; 12:biom12121902. [PMID: 36551330 PMCID: PMC9775183 DOI: 10.3390/biom12121902] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer metabolic reprogramming is essential for maintaining cancer cell survival and rapid replication. A common target of this metabolic reprogramming is one-carbon metabolism which is notable for its function in DNA synthesis, protein and DNA methylation, and antioxidant production. Polyamines are a key output of one-carbon metabolism with widespread effects on gene expression and signaling. As a result of these functions, one-carbon and polyamine metabolism have recently drawn a lot of interest for their part in cancer malignancy. Therapeutic inhibitors that target one-carbon and polyamine metabolism have thus been trialed as anticancer medications. The significance and future possibilities of one-carbon and polyamine metabolism as a target in cancer therapy are discussed in this review.
Collapse
Affiliation(s)
- Anowarul Islam
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Zeeshan Shaukat
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Rashid Hussain
- Clinical and Health Sciences, University of South Australia, Adelaide 5001, Australia
| | - Stephen L. Gregory
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
- Correspondence: ; Tel.: +61-0466987583
| |
Collapse
|
13
|
Stokke C, Nørgaard JN, Feiring Phillips H, Sherwani A, Nuruddin S, Connelly J, Schjesvold F, Revheim ME. Comparison of [ 18F]fluciclovine and [ 18F]FDG PET/CT in Newly Diagnosed Multiple Myeloma Patients. Mol Imaging Biol 2022; 24:842-851. [PMID: 35501622 PMCID: PMC9581841 DOI: 10.1007/s11307-022-01734-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 10/27/2022]
Abstract
PURPOSE [18F]FDG PET/CT in multiple myeloma (MM) is currently the best technology to demonstrate patchy and extramedullary disease. However, [18F]FDG PET has some limitations, and imaging with alternative tracers should be explored. In this study, we aimed to evaluate the performance of [18F]fluciclovine PET compared to [18F]FDG PET in newly diagnosed MM patients. PROCEDURES Thirteen newly diagnosed transplant eligible MM patients were imaged both with [18F]FDG PET/CT and [18F]fluciclovine PET/CT within 1 week in a prospective study. The subjects were visually assessed positive or negative for disease. The number of lesions and the SUVmax of selected lesions were measured for both tracers. Furthermore, tracer uptake ratios were obtained by dividing lesion SUVmax by blood or bone marrow SUVmax. Between-group differences and correlations were assessed with paired t-tests and Pearson tests. Bone marrow SUVs were compared to bone marrow plasma cell percentage in biopsy samples. RESULTS Nine subjects were assessed positively by [18F]FDG PET (69%) and 12 positives by [18F]fluciclovine PET (92%). All positive subjects had [18F]fluciclovine scans that were qualitatively scored as easier to interpret visually than the [18F]FDG scans. The number of lesions was also higher; seven of nine subjects with distinct hot spots on [18F]fluciclovine PET had fewer or no visible lesions on [18F]FDG PET. The mean lesion SUVmax values were 8.2 and 3.8 for [18F]fluciclovine and [18F]FDG, respectively. The mean tumour to blood values were 6.4 and 2.0 for [18F]fluciclovine and [18F]FDG, and the mean ratios between tumour and bone marrow were 2.1 and 1.5 for [18F]fluciclovine and [18F]FDG. The lesion SUVmax and ratios were significantly higher for [18F]fluciclovine (all p < 0.01). Local [18F]fluciclovine SUVmax or SUVmean values in os ilium and the percentage of plasma cells in bone marrow biopsies were linearly correlated (p = 0.048). There were no significant correlations between [18F]FDG SUVs and plasma cells (p = 0.82). CONCLUSIONS Based on this pilot study, [18F]fluciclovine is a promising tracer for MM. The visual and semi-quantitative evaluations indicate that [18F]fluciclovine PET/CT can out-perform [18F]FDG PET/CT at diagnosis.
Collapse
Affiliation(s)
- Caroline Stokke
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.
- Dep. of Physics, University of Oslo, Oslo, Norway.
| | - Jakob Nordberg Nørgaard
- Oslo Myeloma Center, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Alexander Sherwani
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | | | - James Connelly
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Oslo University Hospital, Oslo, Norway
- KG Jebsen Center for B Cell Malignancies, University of Oslo, Oslo, Norway
| | - Mona-Elisabeth Revheim
- Division for Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Morales-Lozano MI, Rodriguez-Otero P, Sancho L, Nuñez-Cordoba JM, Prieto E, Marcos-Jubilar M, Rosales JJ, Alfonso A, Guillen EF, San-Miguel J, Garcia-Velloso MJ. 11C-Methionine PET/CT in Assessment of Multiple Myeloma Patients: Comparison to 18F-FDG PET/CT and Prognostic Value. Int J Mol Sci 2022; 23:ijms23179895. [PMID: 36077292 PMCID: PMC9456410 DOI: 10.3390/ijms23179895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/11/2022] [Accepted: 08/24/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy and remains incurable despite therapeutic advances. 18F-FDG (FDG) PET/CT is a relevant tool MM for staging and it is the reference imaging technique for treatment evaluation. However, it has limitations, and investigation of other PET tracers is required. Preliminary results with L-methyl-[11C]- methionine (MET), suggest higher sensitivity than 18F-FDG. This study aimed to compare the diagnostic accuracy and prognostic value of 1FDG and MET in MM patients. We prospectively compared FDG and MET PET/CT for assessment of bone disease and extramedullary disease (EMD) in a series of 52 consecutive patients (8 smoldering MM, 18 newly diagnosed MM and 26 relapsed MM patients). Bone marrow (BM) uptake patterns and the detection of focal lesions (FLs) and EMD were compared. Furthermore, FDG PET parameters with known MM prognostic value were explored for both tracers, as well as total lesion MET uptake (TLMU). Median patient age was 61 years (range, 37–83 years), 54% were male, 13% of them were in stage ISS (International Staging System) III, and 31% had high-risk cytogenetics. FDG PET/CT did not detect active disease in 6 patients, while they were shown to be positive by MET PET/CT. Additionally, MET PET/CT identified a higher number of FLs than FDG in more than half of the patients (63%). For prognostication we focussed on the relapsed cohort, due to the low number of progressions in the two other cohorts. Upon using FDG PET/CT in relapsed patients, the presence of more than 3 FLs (HR 4.61, p = 0.056), more than 10 FLs (HR 5.65, p = 0.013), total metabolic tumor volume (TMTV) p50 (HR 4.91, p = 0.049) or TMTV p75 (HR 5.32, p = 0.016) were associated with adverse prognosis. In MET PET/CT analysis, TMTV p50 (HR 4.71, p = 0.056), TMTV p75 (HR 6.27, p = 0.007), TLMU p50 (HR 8.8, p = 0.04) and TLMU p75 (HR 6.3, p = 0.007) adversely affected PFS. This study confirmed the diagnostic and prognostic value of FDG in MM. In addition, it highlights that MET has higher sensitivity than FDG PET/CT for detection of myeloma lesions, including FLs. Moreover, we show, for the first time, the prognostic value of TMTV and TLMU MET PET/CT in the imaging evaluation of MM patients.
Collapse
Affiliation(s)
- Maria I. Morales-Lozano
- Department of Nuclear Medicine, Clinica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Paula Rodriguez-Otero
- Department of Haematology, Clínica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Lidia Sancho
- Department of Nuclear Medicine, Clinica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Jorge M. Nuñez-Cordoba
- Research Support Service, Central Clinical Trials Unit, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Elena Prieto
- Department of Nuclear Medicine, Clinica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Maria Marcos-Jubilar
- Department of Haematology, Clínica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Juan J. Rosales
- Department of Nuclear Medicine, Clinica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Ana Alfonso
- Department of Haematology, Clínica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Edgar F. Guillen
- Department of Nuclear Medicine, Clinica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Jesus San-Miguel
- Department of Haematology, Clínica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Maria J. Garcia-Velloso
- Department of Nuclear Medicine, Clinica Universidad de Navarra, CCUN Applied Medical Research, Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
- Correspondence:
| |
Collapse
|
15
|
Zhang S, Shang J, Ye W, Zhao T, Xu H, Zeng H, Wang L. Recent developments on the application of molecular probes in multiple myeloma: Beyond [18F]FDG. Front Bioeng Biotechnol 2022; 10:920882. [PMID: 36091426 PMCID: PMC9459033 DOI: 10.3389/fbioe.2022.920882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is a neoplastic plasma cell proliferative disorder characterized by various osteolytic bone destruction as a radiological morphological marker. Functional imaging, particularly nuclear medicine imaging, is a promising method to visualize disease processes before the appearance of structural changes by targeting specific biomarkers related to metabolism ability, tumor microenvironment as well as neoplastic receptors. In addition, by targeting particular antigens with therapeutic antibodies, immuno-PET imaging can support the development of personalized theranostics. At present, various imaging agents have been prepared and evaluated in MM at preclinical and clinical levels. A summary overview of molecular functional imaging in MM is provided, and commonly used radiotracers are characterized.
Collapse
Affiliation(s)
- Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingjie Shang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Tianming Zhao
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Hui Zeng, ; Lu Wang,
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Hui Zeng, ; Lu Wang,
| |
Collapse
|
16
|
Heidemeier A, Schloetelburg W, Thurner A, Metz C, Heidemeier H, Rasche L, Martin Kortuem K, Boeckle D, Weiland E, Benkert T, Nickel D, Werner R, Buck AK, Bley TA. Multi-parametric whole-body MRI evaluation discerns vital from non-vital multiple myeloma lesions as validated by 18F-FDG and 11C-methionine PET/CT. Eur J Radiol 2022; 155:110493. [PMID: 36027759 DOI: 10.1016/j.ejrad.2022.110493] [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: 12/29/2021] [Revised: 08/04/2022] [Accepted: 08/15/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE We tested a novel multi-parametric (mp) whole body (WB)-MRI evaluation algorithm for medullary lesions in comparison to positron emission tomography (PET) radiotracers 18F-fluorodeoxyglucose (18F-FDG) and 11C-methionine (11C-MET). METHODS AND MATERIALS This retrospective single-center study included 44 MM patients, who received both 18F-FDG-PET and WB-MRI within ten days. MRI classified focal lesions as vital when showing 1) significant diffusion-restriction, 2) a fat fraction (FF) less than 20 % and 3) homogenous hypointensity on T2-weighted images. On a lesion-by-lesion level the findings were compared to 18F-FDG PET by using a 5-point scoring system (analogous to the Deauville score [DS]). In 24/44 (55 %) patients additional comparison to 11C-MET PET was available. RESULTS Among two radiologists, an excellent inter-observer reliability for mpWB-MRI in a total of 84 medullary lesions was observed (ICC = 1, k = 1, p <.01). 16/17 (94.1 %) MRI-classified vital lesions had a DS of 4 or 5 on either 18F-FDG-PET or 11C-MET-PET. MRI-rated non-vital lesions correlated with PET-based DS ≤ 3. When results of mpWB-MRI were compared to 18F-FDG, a fair inter-observer agreement was recorded (ICC = 0.52, k = 0.53, p <.01), while for 11C-MET, an excellent concordance rate was achieved (ICC = 0.81, k = 0.79, p <.01). CONCLUSION The proposed mpWB-MRI interpretation algorithm allowed to assess tumor activity of myeloma lesions with high inter-observer reproducibility. We observed a substantial concordance between the mpWB-MRI classification of lesions and PET assessment based on a semi-automatically calculated 5-point scoring system analogous to the Deauville scores.
Collapse
Affiliation(s)
- Anke Heidemeier
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany.
| | - Wiebke Schloetelburg
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - Annette Thurner
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - Corona Metz
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - Heike Heidemeier
- Department of Psychology, Private University Göttingen, Weender Landstrasse 3-7, 37073 Göttingen, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - K Martin Kortuem
- Department of Internal Medicine II, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - David Boeckle
- Department of Internal Medicine II, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - Elisabeth Weiland
- Department of MR Application Predevelopment, Siemens Healthcare, 91301 Erlangen, Germany
| | - Thomas Benkert
- Department of MR Application Predevelopment, Siemens Healthcare, 91301 Erlangen, Germany
| | - Dominik Nickel
- Department of MR Application Predevelopment, Siemens Healthcare, 91301 Erlangen, Germany
| | - Rudolf Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - Andreas Konrad Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| | - Thorsten Alexander Bley
- Department of Diagnostic and Interventional Radiology, University Hospital Würzburg, Oberdürrbacherstr 6, 97080 Würzburg, Germany
| |
Collapse
|
17
|
Kraus S, Klassen P, Kircher M, Dierks A, Habringer S, Gäble A, Kortüm KM, Weinhold N, Ademaj-Kospiri V, Werner RA, Schirbel A, Buck AK, Herhaus P, Wester HJ, Rosenwald A, Weber WA, Einsele H, Keller U, Rasche L, Lapa C. Reduced splenic uptake on 68Ga-Pentixafor-PET/CT imaging in multiple myeloma - a potential imaging biomarker for disease prognosis. Am J Cancer Res 2022; 12:5986-5994. [PMID: 35966583 PMCID: PMC9373803 DOI: 10.7150/thno.75847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/24/2022] [Indexed: 11/18/2022] Open
Abstract
Beyond being a key factor for tumor growth and metastasis in human cancer, C-X-C motif chemokine receptor 4 (CXCR4) is also highly expressed by a number of immune cells, allowing for non-invasive read-out of inflammatory activity. With two recent studies reporting on prognostic implications of the spleen signal in diffusion-weighted magnetic resonance imaging in patients with plasma cell dyscrasias, the aim of this study was to correlate splenic 68Ga-Pentixafor uptake in multiple myeloma (MM) with clinical parameters and to evaluate its prognostic impact. Methods: Eighty-seven MM patients underwent molecular imaging with 68Ga-Pentixafor-PET/CT. Splenic CXCR4 expression was semi-quantitatively assessed by peak standardized uptake values (SUVpeak) and corresponding spleen-to-bloodpool ratios (TBR) and correlated with clinical and prognostic features as well as survival parameters. Results:68Ga-Pentixafor-PET/CT was visually positive in all MM patients with markedly heterogeneous tracer uptake in the spleen. CXCR4 expression determined by 68Ga-Pentixafor-PET/CT corresponded with advanced disease and was inversely associated with the number of previous treatment lines as compared to controls or untreated smouldering multiple myeloma patients (SUVpeakSpleen 4.06 ± 1.43 vs. 6.02 ± 1.16 vs. 7.33 ± 1.40; P < 0.001). Moreover, reduced splenic 68Ga-Pentixafor uptake was linked to unfavorable clinical outcome. Patients with a low SUVpeakSpleen (<3.35) experienced a significantly shorter overall survival of 5 months as compared to 62 months in patients with a high SUVpeakSpleen >5.79 (P < 0.001). Multivariate Cox analysis confirmed SUVpeakSpleen as an independent predictor of survival (HR 0.75; P = 0.009). Conclusion: These data suggest that splenic 68Ga-Pentixafor uptake might provide prognostic information in pre-treated MM patients similar to what was reported for diffusion-weighted magnetic resonance imaging. Further research to elucidate the underlying biologic implications is warranted.
Collapse
Affiliation(s)
- Sabrina Kraus
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Philipp Klassen
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Alexander Dierks
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Stefan Habringer
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alexander Gäble
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Klaus Martin Kortüm
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Niels Weinhold
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Valëza Ademaj-Kospiri
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany.,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Peter Herhaus
- Technical University Munich, School of Medicine, Klinikum rechts der Isar, Clinic and Policlinic for Internal Medicine III, Munich, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University of Munich, Munich, Germany
| | | | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
18
|
Abstract
Multiple myeloma (MM) accounts for 0.9% of cancer diagnoses, and incidence and mortality rate have increased in previous years. 18F-fluorodeoxyglucose (FDG) PET-computed tomography (CT) is an established modality for MM evaluation. MR imaging is helpful where 18F-FDG PET-CT is lacking. To standardize PET reporting, methods like Italian Myeloma Criteria for PET Use and Deauville criteria have been studied. Tracers like 11C-acetate and 11C-choline/18F-fluoromethylcholine (FCH) have shown higher sensitivity and detected more focal lesions and diffuse involvement than 18F-FDG PET-CT. 18F-FCH showed higher maximum standardized uptake value than 18FDG. 11C-methionine appears to be the best radiopharmaceutical, apart from 18F-FDG, for evaluating MM.
Collapse
Affiliation(s)
- Angel Hemrom
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Avinash Tupalli
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Abass Alavi
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Rakesh Kumar
- Diagnostic Nuclear Medicine Division, Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
19
|
Wenz J, Arndt F, Samnick S. A new concept for the production of 11C-labelled radiotracers. EJNMMI Radiopharm Chem 2022; 7:6. [PMID: 35347490 PMCID: PMC8960519 DOI: 10.1186/s41181-022-00159-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/09/2022] [Indexed: 11/10/2022] Open
Abstract
Background The GMP-compliant production of radiopharmaceuticals has been performed using disposable units (cassettes) with a dedicated synthesis module. To expand this “plug ‘n’ synthesize” principle to a broader scope of modules we developed a pressure controlled setup that offers an alternative to the usual stepper motor controlled rotary valves. The new concept was successfully applied to the synthesis of N-methyl-[11C]choline, L-S-methyl-[11C]methionine and [11C]acetate. Results The target gas purification of cyclotron produced [11C]CO2 and subsequent conversion to [11C]MeI was carried out on a TRACERlab Fx C Pro module. The labelling reactions were controlled with a TRACERlab Fx FE module. With the presented modular principle we were able to produce N-methyl-[11C]choline and L-S-methyl-[11C]methionine by loading a reaction loop with neat N,N'-dimethylaminoethanol (DMAE) or an ethanol/water mixture of NaOH and L-homocysteine (L-HC), respectively and a subsequent reaction with [11C]MeI. After 18 min N-methyl-[11C]choline was isolated with 52% decay corrected yield and a radiochemical purity of > 99%. For L-S-methyl-[11C]methionine the total reaction time was 19 min reaction, yielding 25% of pure product (> 97%). The reactor design was used as an exemplary model for the technically challenging [11C]acetate synthesis. The disposable unit was filled with 1 mL MeMgCl (0.75 M) in tetrahydrofuran (THF) bevore [11C]CO2 was passed through. After complete release of [11C]CO2 the reaction mixture was quenched with water and guided through a series of ion exchangers (H+, Ag+ and OH−). The product was retained on a strong anion exchanger, washed with water and finally extracted with saline. The product mixture was acidified and degassed to separate excess [11C]CO2 before dispensing. Under these conditions the total reaction time was 18 ± 2 min and pure [11C]acetate (n = 10) was isolated with a decay corrected yield of 51 ± 5%. Conclusion Herein, we described a novel single use unit for the synthesis of carbon-11 labelled tracers for preclinical and clinical applications of N-methyl-[11C]choline, L-S-methyl-[11C]methionine and [11C]acetate. Supplementary Information The online version contains supplementary material available at 10.1186/s41181-022-00159-y.
Collapse
Affiliation(s)
- Jan Wenz
- Department of Nuclear Medicine, Interdisciplinary PET Center, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany.
| | - Felix Arndt
- Department of Nuclear Medicine, Interdisciplinary PET Center, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, Interdisciplinary PET Center, Universitätsklinikum Würzburg, Oberdürrbacher Strasse 6, 97080, Würzburg, Germany
| |
Collapse
|
20
|
von Hinten J, Kircher M, Dierks A, Pfob CH, Higuchi T, Pomper MG, Rowe SP, Buck AK, Samnick S, Werner RA, Lapa C. Molecular Imaging in Multiple Myeloma-Novel PET Radiotracers Improve Patient Management and Guide Therapy. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:801792. [PMID: 39354963 PMCID: PMC11440847 DOI: 10.3389/fnume.2022.801792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/01/2022] [Indexed: 10/03/2024]
Abstract
Due to its proven value in imaging of multiple myeloma (MM), including staging, prognostication, and assessment of therapy response, 2-deoxy-2-[18F]fluoro-D-glucose (FDG) positron emission tomography (PET) is utilized extensively in the clinic. However, its accuracy is hampered by imperfect sensitivity (e.g., so-called FDG-negative MM) as well as specificity (e.g., inflammatory processes), with common pitfalls including fractures and degenerative changes. Novel approaches providing a read-out of increased protein or lipid membrane syntheses, such as [11C]methionine and [11C]choline or the C-X-C motif chemokine receptor 4-targeting radiotracer [68Ga]Pentixafor, have already been shown to be suitable adjuncts or alternatives to FDG. In the present focused review, those imaging agents along with their theranostic potential in the context of MM are highlighted.
Collapse
Affiliation(s)
- Johannes von Hinten
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Alexander Dierks
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Christian H. Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf A. Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| |
Collapse
|
21
|
Mesguich C, Hulin C, Latrabe V, Lascaux A, Bordenave L, Hindié E. 18 F-FDG PET/CT and MRI in the Management of Multiple Myeloma: A Comparative Review. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 1:808627. [PMID: 39355637 PMCID: PMC11440970 DOI: 10.3389/fnume.2021.808627] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/27/2021] [Indexed: 10/03/2024]
Abstract
During the last two decades, the imaging landscape of multiple myeloma (MM) has evolved with whole-body imaging techniques such as fluorodeoxyglucose positron emission tomography-computed tomography (18F-FDG PET/CT) and MRI replacing X-ray skeletal survey. Both imaging modalities have high diagnostic performance at the initial diagnosis of MM and are key players in the identification of patients needing treatment. Diffusion-weighted MRI has a high sensitivity for bone involvement, while 18F-FDG PET/CT baseline parameters carry a strong prognostic value. The advent of more efficient therapeutics, such as immunomodulatory drugs and proteasome inhibitors, has called for the use of sensitive imaging techniques for monitoring response to treatment. Diffusion-weighted MRI could improve the specificity of MRI for tumor response evaluation, but questions remain regarding its role as a prognostic factor. Performed at key time points of treatment in newly diagnosed MM patients, 18F-FDG PET/CT showed a strong association with relapse risk and survival. The deployment of minimal residual disease detection at the cellular or the molecular level may raise questions on the role of these imaging techniques, which will be addressed. This review summarizes and outlines the specificities and respective roles of MRI and 18F-FDG PET/CT in the management of MM.
Collapse
Affiliation(s)
- Charles Mesguich
- Department of Nuclear Medicine, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
- University of Bordeaux, IMB, UMR CNRS 5251, INRIA Project Team Monc, Talence, France
| | - Cyrille Hulin
- Department of Haematology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Valérie Latrabe
- Department of Radiology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Axelle Lascaux
- Department of Haematology, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Laurence Bordenave
- Department of Nuclear Medicine, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Elif Hindié
- Department of Nuclear Medicine, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
- University of Bordeaux, INCIA UMR-CNRS 5287, Talence, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
22
|
Morath V, Heider M, Mittelhäuser M, Rolbieski H, Stroh J, Calais J, Eiber M, Bassermann F, Weber WA. Initial evaluation of [ 18F]-FACBC for PET imaging of multiple myeloma. EJNMMI Res 2022; 12:4. [PMID: 35099620 PMCID: PMC8804025 DOI: 10.1186/s13550-022-00876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022] Open
Abstract
RATIONALE Multiple myeloma (MM) cells synthesize large amounts of paraproteins, making radiolabeled amino acids promising candidates for PET imaging of MM patients. METHODS We compare tumor uptake of the two amino acid analogs [18F]-fluoroethyltyrosine and [18F]-FACBC in a MM xenograft model and show the feasibility of PET imaging with [18F]-FACBC in a MM patient. RESULTS Preclinically [18F]-FACBC showed superior performance, mainly due to the uptake via the ASC-system. In a subsequent proof-of-concept investigation [18F]-FACBC PET was performed in a MM patient. It allowed identification of both lesions with and without CT correlate (SUVmean 8.0 or 7.9) based on higher uptake compared to normal bone marrow (SUVmean 5.7). Bone signal was elevated compared to non-MM patients, and, thus [18F]-FACBC potentially allows the assessment of bone marrow infiltration. CONCLUSION The FDA/EMA approved PET agent [18F]-FACBC is promising for imaging MM and should be further evaluated in prospective clinical studies.
Collapse
Affiliation(s)
- Volker Morath
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Michael Heider
- Department of Medicine III, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Markus Mittelhäuser
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Hannes Rolbieski
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Jacob Stroh
- Department of Medicine III, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Jérémie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Florian Bassermann
- Department of Medicine III, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, 81675, Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, 81675, Munich, Germany.
| |
Collapse
|
23
|
Minamimoto R. Amino Acid and Proliferation PET/CT for the Diagnosis of Multiple Myeloma. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 1:796357. [PMID: 39355641 PMCID: PMC11440849 DOI: 10.3389/fnume.2021.796357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/22/2021] [Indexed: 10/03/2024]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by infiltration of monoclonal plasma cells in the bone marrow (BM). The standard examination performed for the assessment of bone lesions has progressed from radiographic skeletal survey to the more advanced imaging modalities of computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography/computed tomography (PET/CT). The Durie-Salmon PLUS staging system (upgraded from the Durie-Salmon staging system) applies 2-[18F]-fluoro-2-deoxy-glucose (18F-FDG) PET/CT, and MRI findings to the staging of MM, and 18F-FDG PET/CT has been incorporated into the International Myeloma Working Group (IMWG) guidelines for the diagnosis and staging of MM. However, 18F-FDG PET/CT has significant limitations in the assessment of diffuse BM infiltration and in the differentiation of MM lesions from inflammatory or infectious lesions. The potential of several new PET tracers that exploit the underlying disease mechanism of MM has been evaluated in terms of improving the diagnosis. L-type amino acid transporter 1 (LAT1), a membrane protein that transports neutral amino acids, is associated with cell proliferation and has strong ability to represent the status of MM. This review evaluates the potential of amino acid and proliferation PET tracers for diagnosis and compares the characteristics and accuracy of non-FDG tracers in the management of patients with MM.
Collapse
Affiliation(s)
- Ryogo Minamimoto
- Division of Nuclear Medicine, Department of Radiology, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Gonçalves AC, Richiardone E, Jorge J, Polónia B, Xavier CPR, Salaroglio IC, Riganti C, Vasconcelos MH, Corbet C, Sarmento-Ribeiro AB. Impact of cancer metabolism on therapy resistance - Clinical implications. Drug Resist Updat 2021; 59:100797. [PMID: 34955385 DOI: 10.1016/j.drup.2021.100797] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite an increasing arsenal of anticancer therapies, many patients continue to have poor outcomes due to the therapeutic failures and tumor relapses. Indeed, the clinical efficacy of anticancer therapies is markedly limited by intrinsic and/or acquired resistance mechanisms that can occur in any tumor type and with any treatment. Thus, there is an urgent clinical need to implement fundamental changes in the tumor treatment paradigm by the development of new experimental strategies that can help to predict the occurrence of clinical drug resistance and to identify alternative therapeutic options. Apart from mutation-driven resistance mechanisms, tumor microenvironment (TME) conditions generate an intratumoral phenotypic heterogeneity that supports disease progression and dismal outcomes. Tumor cell metabolism is a prototypical example of dynamic, heterogeneous, and adaptive phenotypic trait, resulting from the combination of intrinsic [(epi)genetic changes, tissue of origin and differentiation dependency] and extrinsic (oxygen and nutrient availability, metabolic interactions within the TME) factors, enabling cancer cells to survive, metastasize and develop resistance to anticancer therapies. In this review, we summarize the current knowledge regarding metabolism-based mechanisms conferring adaptive resistance to chemo-, radio-and immunotherapies as well as targeted therapies. Furthermore, we report the role of TME-mediated intratumoral metabolic heterogeneity in therapy resistance and how adaptations in amino acid, glucose, and lipid metabolism support the growth of therapy-resistant cancers and/or cellular subpopulations. We also report the intricate interplay between tumor signaling and metabolic pathways in cancer cells and discuss how manipulating key metabolic enzymes and/or providing dietary changes may help to eradicate relapse-sustaining cancer cells. Finally, in the current era of personalized medicine, we describe the strategies that may be applied to implement metabolic profiling for tumor imaging, biomarker identification, selection of tailored treatments and monitoring therapy response during the clinical management of cancer patients.
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium
| | - Joana Jorge
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Bárbara Polónia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | | | - Chiara Riganti
- Department of Oncology, School of Medicine, University of Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy of the University of Porto, Porto, Portugal
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Belgium.
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH) and University Clinic of Hematology, Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR) - Group of Environment Genetics and Oncobiology (CIMAGO), FMUC, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Hematology Service, Centro Hospitalar e Universitário de Coimbra (CHUC), Coimbra, Portugal.
| |
Collapse
|
25
|
Spinnato P, Filonzi G, Conficoni A, Facchini G, Ponti F, Sambri A, De Paolis M, Cavo M, Salizzoni E, Nanni C. Skeletal Survey in Multiple Myeloma: Role of Imaging. Curr Med Imaging 2021; 17:956-965. [PMID: 33573573 DOI: 10.2174/1573405617666210126155129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/23/2020] [Accepted: 12/18/2020] [Indexed: 11/22/2022]
Abstract
Bone disease is the hallmark of multiple myeloma. Skeletal lesions are evaluated to establish the diagnosis, to choose the therapies and also to assess the response to treatments. Due to this, imaging procedures play a key role in the management of multiple myeloma. For decades, conventional radiography has been the standard imaging modality. Subsequently, advances in the treatment of multiple myeloma have increased the need for an accurate evaluation of skeletal disease. The introduction of new high performant imaging tools, such as whole-body lowdose computed tomography, different types of magnetic resonance imaging studies, and 18F-fluorodeoxyglucose positron emission tomography, replaced the conventional radiography. In this review, we analyze the diagnostic potentials, indications of use, and applications of the imaging tools nowadays available. Whole-body low-dose CT should be considered as the imaging modality of choice for the initial assessment of multiple myeloma lytic bone lesions. MRI is the gold-standard for the detection of bone marrow involvement, while PET/CT is the preferred technique in the assessment of response to therapy. Both MRI and PET/CT are able to provide prognostic information.
Collapse
Affiliation(s)
- Paolo Spinnato
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Alberto Conficoni
- Neuroradiology Unit, Department of Radiology, Azienda Ospedaliero-Universitaria di Ferrara, Ferrara, Italy
| | - Giancarlo Facchini
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Federico Ponti
- Diagnostic and Interventional Radiology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Andrea Sambri
- Orthopaedic Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Michele Cavo
- "Seràgnoli" Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Eugenio Salizzoni
- Imaging Division, Clinical Department of Radiological and Histocytopathological Sciences, University of Bologna, Sant'Orsola-Malpighi Hospital, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine, MNM AOU S.Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
26
|
Abstract
The use of PET imaging agents in oncology, cardiovascular disease, and neurodegenerative disease shows the power of this technique in evaluating the molecular and biological characteristics of numerous diseases. These agents provide crucial information for designing therapeutic strategies for individual patients. Novel PET tracers are in continual development and many have potential use in clinical and research settings. This article discusses the potential applications of tracers in diagnostics, the biological characteristics of diseases, the ability to provide prognostic indicators, and using this information to guide treatment strategies including monitoring treatment efficacy in real time to improve outcomes and survival.
Collapse
|
27
|
Zanoni L, Mattana F, Calabrò D, Paccagnella A, Broccoli A, Nanni C, Fanti S. Overview and recent advances in PET/CT imaging in lymphoma and multiple myeloma. Eur J Radiol 2021; 141:109793. [PMID: 34148014 DOI: 10.1016/j.ejrad.2021.109793] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/18/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023]
Abstract
Imaging in hematological diseases has evolved extensively over the past several decades. Positron emission tomography/computed tomography (PET/CT) with of 2-[18 F]-fluoro-2-deoxy-d-glucose ([18 F] FDG) is currently essential for accurate staging and for early and late therapy response assessment for all FDG-avid lymphoproliferative histologies. The widely adopted visual Deauville 5-point scale and Lugano Classification recommendations have recently standardized PET scans interpretation and improved lymphoma patient management. In addition [18 F] FDG-PET is routinely recommended for initial evaluation and treatment response assessment of Multiple Myeloma (MM) with significant contribution in risk-stratification and prognostication, although magnetic resonance imaging remains the Gold Standard for the assessment of bone marrow involvement. In this review, an overview of the role of [18 F] FDG-PET, in hematological malignancies is provided, particularly focusing on Hodgkin lymphoma (HL) and Diffuse Large B Cell Lymphoma (DLBCL), both in adult and pediatric populations, and MM, at each point of patient management. Potential alternative molecular imaging applications in this field, such as non-[18 F] FDG-tracers, whole body magnetic resonance imaging (WB-MRI), hybrid PET/MRI and emerging radiomics research are briefly presented.
Collapse
Affiliation(s)
- Lucia Zanoni
- IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Nuclear Medicine, via Massarenti 9, 40138, Bologna, Italy.
| | - Francesco Mattana
- Nuclear Medicine, DIMES, Alma Mater studiorum, Università di Bologna, Bologna, Italy.
| | - Diletta Calabrò
- Nuclear Medicine, DIMES, Alma Mater studiorum, Università di Bologna, Bologna, Italy.
| | - Andrea Paccagnella
- Nuclear Medicine, DIMES, Alma Mater studiorum, Università di Bologna, Bologna, Italy.
| | - Alessandro Broccoli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy; Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy.
| | - Cristina Nanni
- IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Nuclear Medicine, via Massarenti 9, 40138, Bologna, Italy.
| | - Stefano Fanti
- IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Nuclear Medicine, via Massarenti 9, 40138, Bologna, Italy; Nuclear Medicine, DIMES, Alma Mater studiorum, Università di Bologna, Bologna, Italy.
| |
Collapse
|
28
|
Bertamini L, D'Agostino M, Gay F. MRD Assessment in Multiple Myeloma: Progress and Challenges. Curr Hematol Malig Rep 2021; 16:162-171. [PMID: 33950462 DOI: 10.1007/s11899-021-00633-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Over the last decade, the development of effective treatment approaches for multiple myeloma (MM) has been associated with higher response rates and longer survival. In patients who achieve complete response, several high sensitivity techniques have been studied to assess minimal residual disease (MRD) and detect residual neoplastic cells within the bone marrow (by flow cytometry or molecular biology techniques) or outside the bone marrow (by imaging or circulating disease markers in the peripheral blood). This is of utmost importance, since residual disease can drive clinical relapse. This review focuses on the progress made in the assessment of MRD in MM. RECENT FINDINGS The achievement of MRD negativity after therapy is considered prognostically important for MM patients, and data from clinical trials and meta-analyses have confirmed that it is strongly associated with better survival. Along with well-known techniques, such as next-generation sequencing (NGS), next-generation flow (NGF), and positron emission tomography/computed tomography (PET/CT), other methods such as mass spectrometry (MS) and circulating tumor cells are under study. Intensive treatment regimens at diagnosis can lead up to 70% of MRD negativity in MM patients, although the current proportion of curable patients is still unknown. Today, clinicians who treat MM deal with MRD assessment in routine clinical practice. Its appropriate use in therapeutic decision making may be the most fascinating and challenging issue to be addressed over the next few years.
Collapse
Affiliation(s)
- Luca Bertamini
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, via Genova 3, 10126, Torino, Italy
| | - Mattia D'Agostino
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, via Genova 3, 10126, Torino, Italy
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, via Genova 3, 10126, Torino, Italy.
| |
Collapse
|
29
|
Ultra-low dose whole-body CT for attenuation correction in a dual tracer PET/CT protocol for multiple myeloma. Phys Med 2021; 84:1-9. [PMID: 33799056 DOI: 10.1016/j.ejmp.2021.03.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/22/2021] [Accepted: 03/13/2021] [Indexed: 01/17/2023] Open
Abstract
PURPOSE To investigate within phantoms the minimum CT dose allowed for accurate attenuation correction of PET data and to quantify the effective dose reduction when a CT for this purpose is incorporated in the clinical setting. METHODS The NEMA image quality phantom was scanned within a large parallelepiped container. Twenty-one different CT images were acquired to correct attenuation of PET raw data. Radiation dose and image quality were evaluated. Thirty-one patients with proven multiple myeloma who underwent a dual tracer PET/CT scan were retrospectively reviewed. 18F-fluorodeoxyglucose PET/CT included a diagnostic whole-body low dose CT (WBLDCT: 120 kV-80mAs) and 11C-Methionine PET/CT included a whole-body ultra-low dose CT (WBULDCT) for attenuation correction (100 kV-40mAs). Effective dose and image quality were analysed. RESULTS Only the two lowest radiation dose conditions (80 kV-20mAs and 80 kV-10mAs) produced artifacts in CT images that degraded corrected PET images. For all the other conditions (CTDIvol ≥ 0.43 mGy), PET contrast recovery coefficients varied less than ± 1.2%. Patients received a median dose of 6.4 mSv from diagnostic CT and 2.1 mSv from the attenuation correction CT. Despite the worse image quality of this CT, 94.8% of bone lesions were identifiable. CONCLUSION Phantom experiments showed that an ultra-low dose CT can be implemented in PET/CT procedures without any noticeable degradation in the attenuation corrected PET scan. The replacement of the standard CT for this ultra-low dose CT in clinical PET/CT scans involves a significant radiation dose reduction.
Collapse
|
30
|
Positron Emission Tomography (PET) Imaging of Multiple Myeloma in a Post-Treatment Setting. Diagnostics (Basel) 2021; 11:diagnostics11020230. [PMID: 33546455 PMCID: PMC7913723 DOI: 10.3390/diagnostics11020230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/23/2021] [Accepted: 01/29/2021] [Indexed: 12/01/2022] Open
Abstract
2-deoxy-2-[18F]fluoro-D-glucose (FDG) positron emission tomography/computed tomography (FDG PET/CT) has an established clinical value in the diagnosis and initial staging of multiple myeloma (MM). In the last ten years, a vast body of literature has shown that this tool can also be of high relevance for monitoring therapy responses, making it the recommended imaging approach in this field. Starting from the strengths and weaknesses of radiological imaging in MM, the present review aims to analyze FDG PET/CT’s current clinical value focusing on therapy response assessment and objective interpretation criteria for therapy monitoring. Given the potential occurrence of patients with MM showing non-FDG-avid bone disease, new opportunities can be provided by non-FDG PET tracers. Accordingly, the potential role of non-FDG PET tracers in this setting has also been discussed.
Collapse
|
31
|
Matteucci F, Paganelli G, Martinelli G, Cerchione C. PET/CT in Multiple Myeloma: Beyond FDG. Front Oncol 2021; 10:622501. [PMID: 33569348 PMCID: PMC7868556 DOI: 10.3389/fonc.2020.622501] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/07/2020] [Indexed: 12/03/2022] Open
Abstract
Recent advances in the diagnosis and treatment of multiple myeloma (MM) have highlighted the importance of imaging methods, not only in the localization and extent of the disease but also in prognostic stratification and assessment of response to therapy. In this context, PET/CT, combining both morphological and functional information, is particularly useful in this pathology. The tracer mostly used is 18F-FDG, a glucose analog, which provides extremely accurate information with a sensitivity ranging from 80 to 100%. However, this tracer has some limitations, mostly related to the physiological uptake of FDG in the bone marrow and brain, which reduce its effectiveness. For this reason, some studies in the literature have evaluated the effectiveness of other PET tracers, which provide information on protein metabolism or the synthesis of metabolic plasma membranes, such as choline and methionine, as well as innovative radiopharmaceuticals, directed against receptors expressed by cells of myeloma, including tracers directed to the chemokine receptor. This review analyzes the characteristics and accuracy of non-FDG tracers in the management of patients with multiple myeloma.
Collapse
Affiliation(s)
- Federica Matteucci
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo studio e la cura dei tumori (IRST) IRCCS, Meldola, Italy
| | - Giovanni Paganelli
- Nuclear Medicine Unit, Istituto Scientifico Romagnolo per lo studio e la cura dei tumori (IRST) IRCCS, Meldola, Italy
| | - Giovanni Martinelli
- Hematology Unit, Istituto Scientifico Romagnolo per lo studio e la cura dei tumori (IRST) IRCCS, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo studio e la cura dei tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
32
|
Maccora D, Caldarella C, Taralli S, Scolozzi V, Lorusso M, Cocciolillo F, Calcagni ML. Diagnostic role of 11C-methionine PET/CT in patients with multiple myeloma and other plasma cell malignancy: a literature review. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-020-00400-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
The Link between Cytogenetics/Genomics and Imaging Patterns of Relapse and Progression in Patients with Relapsed/Refractory Multiple Myeloma: A Pilot Study Utilizing 18F-FDG PET/CT. Cancers (Basel) 2020; 12:cancers12092399. [PMID: 32847053 PMCID: PMC7564496 DOI: 10.3390/cancers12092399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 01/10/2023] Open
Abstract
Utilizing 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT), we performed this pilot study to evaluate the link between cytogenetic/genomic markers and imaging patterns in relapsed/refractory (RR) multiple myeloma (MM). We retrospectively analyzed data of 24 patients with RRMM who were treated at our institution between November 2018 and February 2020. At the last relapse/progression, patients had been treated with a median of three (range 1-10) lines of therapy. Six (25%) patients showed FDG avid extramedullary disease without adjacency to bone. We observed significantly higher maximum standardized uptake values (SUVmax) in patients harboring del(17p) compared with those without del(17p) (p = 0.025). Moreover, a high SUVmax of >15 indicated significantly shortened progression-free survival (PFS) (p = 0.01) and overall survival (OS) (p = 0.0002). One female patient exhibited biallelic TP53 alteration, i.e., deletion and mutation, in whom an extremely high SUVmax of 37.88 was observed. In summary, this pilot study suggested a link between del(17p)/TP53 alteration and high SUVmax on 18F-FDG PET/CT in RRMM patients. Further investigations are highly warranted at this point.
Collapse
|
34
|
Zhou X, Dierks A, Kertels O, Kircher M, Schirbel A, Samnick S, Buck AK, Knorz S, Böckle D, Scheller L, Messerschmidt J, Barakat M, Kortüm KM, Rasche L, Einsele H, Lapa C. 18F-FDG, 11C-Methionine, and 68Ga-Pentixafor PET/CT in Patients with Smoldering Multiple Myeloma: Imaging Pattern and Clinical Features. Cancers (Basel) 2020; 12:cancers12082333. [PMID: 32824832 PMCID: PMC7465161 DOI: 10.3390/cancers12082333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 01/23/2023] Open
Abstract
This study aimed to explore the correlation between imaging patterns and clinical features in patients with smoldering multiple myeloma (SMM) who simultaneously underwent 18F-FDG, 11C-Methionine, and 68Ga-Pentixafor positron emission tomography/computed tomography (PET/CT). We retrieved and analyzed clinical characteristics and PET imaging data of 10 patients with SMM. We found a significant correlation between bone marrow (BM) plasma cell (PC) infiltration and mean standardized uptake values (SUVmean) of lumbar vertebrae L2-L4 on 11C-Methionine PET/CT scans (r = 0.676, p = 0.031) and 68Ga-Pentixafor PET/CT scans (r = 0.839, p = 0.002). However, there was no significant correlation between BM involvement and SUVmean of lumbar vertebrae L2-L4 on 18F-FDG PET/CT scans (r = 0.558, p = 0.093). Similarly, mean target-to-background ratios (TBRmean) of lumbar vertebrae L2-L4 also correlated with bone marrow plasma cell (BMPC) infiltration in 11C-Methionine PET/CT (r = 0.789, p = 0.007) and 68Ga-Pentixafor PET/CT (r = 0.724, p = 0.018) PET/CT. In contrast, we did not observe a significant correlation between BMPC infiltration rate and TBRmean in 18F-FDG PET/CT (r = 0.355, p = 0.313). Additionally, on 11C-Methionine PET/CT scans, we found a significant correlation between BMPC infiltration and TBRmax of lumbar vertebrae L2-L4 (r = 0.642, p = 0.045). In conclusion, 11C-Methionine and 68Ga-Pentixafor PET/CT demonstrate higher sensitivity than 18F-FDG PET/CT in detecting BM involvement in SMM.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Alexander Dierks
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Olivia Kertels
- Department of Diagnostic and Interventional Radiology, University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Malte Kircher
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
| | - Sebastian Knorz
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - David Böckle
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Lukas Scheller
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Janin Messerschmidt
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Mohammad Barakat
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - K. Martin Kortüm
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, 97080 Würzburg, Germany; (X.Z.); (S.K.); (D.B.); (L.S.); (J.M.); (M.B.); (K.M.K.); (L.R.); (H.E.)
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital of Würzburg, 97080 Würzburg, Germany; (A.D.); (M.K.); (A.S.); (S.S.); (A.K.B.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
- Correspondence:
| |
Collapse
|
35
|
Burgos L, Puig N, Cedena MT, Mateos MV, Lahuerta JJ, Paiva B, San-Miguel JF. Measurable residual disease in multiple myeloma: ready for clinical practice? J Hematol Oncol 2020; 13:82. [PMID: 32571377 PMCID: PMC7310444 DOI: 10.1186/s13045-020-00911-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/04/2020] [Indexed: 01/11/2023] Open
Abstract
The landscape of multiple myeloma (MM) has changed considerably in the past two decades regarding new treatments, insight into disease biology and innovation in the techniques available to assess measurable residual disease (MRD) as the most accurate method to evaluate treatment efficacy. The sensitivity and standardization achieved by these techniques together with unprecedented rates of complete remission (CR) induced by new regimens, raised enormous interest in MRD as a surrogate biomarker of patients' outcome and endpoint in clinical trials. By contrast, there is reluctance and general lack of consensus on how to use MRD outside clinical trials. Here, we discuss critical aspects related with the implementation of MRD in clinical practice.
Collapse
Affiliation(s)
- Leire Burgos
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Noemi Puig
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - María-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | | | - Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain
| | - Jesús F San-Miguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), Instituto de Investigación Sanitaria de Navarra (IDISNA), CIBER-ONC number CB16/12/00369, Pamplona, Spain.
| |
Collapse
|
36
|
Can 18F-NaF PET/CT before Autologous Stem Cell Transplantation Predict Survival in Multiple Myeloma? Cancers (Basel) 2020; 12:cancers12051335. [PMID: 32456181 PMCID: PMC7281312 DOI: 10.3390/cancers12051335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Accepted: 05/21/2020] [Indexed: 02/07/2023] Open
Abstract
There is an unmet need for positron emission tomography (PET) radiotracers that can image bone disease in multiple myeloma (MM) in a more sensitive and specific way than the widely used 18F-fluorodeoxyglucose (18F-FDG). Sodium fluoride (18F-NaF) is a highly sensitive tracer of bone reconstruction, evolving as an important imaging agent for the assessment of malignant bone diseases. We attempted to investigate for the first time the prognostic significance of 18F-NaF PET/CT in newly diagnosed, symptomatic MM patients planned for autologous stem cell transplantation (ASCT). Forty-seven patients underwent dynamic and static PET/CT with 18F-NaF before treatment. After correlation with the respective findings on CT and 18F-FDG PET/CT that served as reference, the 18F-NaF PET findings were compared with established factors of high-risk disease, like cytogenetic abnormalities as well as bone marrow plasma cell infiltration rate. Furthermore, the impact of 18F-NaF PET/CT on progression-free survival (PFS) was analyzed. Correlation analysis revealed a moderate, significant correlation of the 18F-NaF parameters SUVaverage and K1 in reference tissue with bone marrow plasma cell infiltration rate. However, no significant correlation was observed regarding all other 18F-NaF PET parameters. Survival analysis revealed that patients with a pathologic 18F-NaF PET/CT have a shorter PFS (median = 36.2 months) than those with a physiologic scan (median = 55.6 months) (p = 0.02). Nevertheless, no quantitative 18F-NaF parameter could be shown to adversely affect PFS. In contrast, the respective analysis for quantitative dynamic 18F-FDG PET/CT revealed that the parameters SUVmax, fractional blood volume (VB), k3 and influx from reference tissue as well as SUVaverage from MM lesions had a significant negative impact on patient survival. The herein presented findings highlight the rather limited role of 18F-NaF PET/CT as a single PET approach in MM.
Collapse
|
37
|
Morales-Lozano MI, Viering O, Samnick S, Rodriguez-Otero P, Buck AK, Marcos-Jubilar M, Rasche L, Prieto E, Kortüm KM, San-Miguel J, Garcia-Velloso MJ, Lapa C. 18F-FDG and 11C-Methionine PET/CT in Newly Diagnosed Multiple Myeloma Patients: Comparison of Volume-Based PET Biomarkers. Cancers (Basel) 2020; 12:cancers12041042. [PMID: 32340251 PMCID: PMC7226577 DOI: 10.3390/cancers12041042] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/02/2022] Open
Abstract
11C-methionine (11C-MET) is a new positron emission tomography (PET) tracer for the assessment of disease activity in multiple myeloma (MM) patients, with preliminary data suggesting higher sensitivity and specificity than 18F-fluorodeoxyglucose (18F-FDG). However, the value of tumor burden biomarkers has yet to be investigated. Our goals were to corroborate the superiority of 11C-MET for MM staging and to compare its suitability for the assessment of metabolic tumor burden biomarkers in comparison to 18F-FDG. Twenty-two patients with newly diagnosed, treatment-naïve symptomatic MM who had undergone 11C-MET and 18F-FDG PET/CT were evaluated. Standardized uptake values (SUV) were determined and compared with total metabolic tumor volume (TMTV) for both tracers: total lesion glycolysis (TLG) and total lesion 11C-MET uptake (TLMU). PET-derived values were compared to Revised International Staging System (R-ISS), cytogenetic, and serologic MM markers such as M component, beta 2 microglobulin (B2M), serum free light chains (FLC), albumin, and lactate dehydrogenase (LDH). In 11 patients (50%), 11C-MET detected more focal lesions (FL) than FDG (p < 0.01). SUVmax, SUVmean, SUVpeak, TMTV, and TLMU were also significantly higher in 11C-MET than in 18F-FDG (p < 0.05, respectively). 11C-MET PET biomarkers had a better correlation with tumor burden (bone marrow plasma cell infiltration, M component; p < 0.05 versus p = n.s. respectively). This pilot study suggests that 11C-MET PET/CT is a more sensitive marker for the assessment of myeloma tumor burden than 18F-FDG. Its implications for prognosis evaluation need further investigation.
Collapse
Affiliation(s)
- Maria I Morales-Lozano
- Department of Nuclear Medicine, University Clinic of Navarra, Center of Applied Medical Research (CIMA), Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain; (M.I.M.-L.); (E.P.)
| | - Oliver Viering
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (O.V.); (S.S.); (A.K.B.); (C.L.)
| | - Samuel Samnick
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (O.V.); (S.S.); (A.K.B.); (C.L.)
| | - Paula Rodriguez-Otero
- Department of Hematology, University Clinic of Navarra, CIMA, CIBERONC, IDISNA, 31008 Pamplona, Spain; (P.R.-O.); (M.M.-J.); (J.S.-M.)
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (O.V.); (S.S.); (A.K.B.); (C.L.)
| | - Maria Marcos-Jubilar
- Department of Hematology, University Clinic of Navarra, CIMA, CIBERONC, IDISNA, 31008 Pamplona, Spain; (P.R.-O.); (M.M.-J.); (J.S.-M.)
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany; (L.R.); (K.M.K.)
| | - Elena Prieto
- Department of Nuclear Medicine, University Clinic of Navarra, Center of Applied Medical Research (CIMA), Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain; (M.I.M.-L.); (E.P.)
| | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany; (L.R.); (K.M.K.)
| | - Jesus San-Miguel
- Department of Hematology, University Clinic of Navarra, CIMA, CIBERONC, IDISNA, 31008 Pamplona, Spain; (P.R.-O.); (M.M.-J.); (J.S.-M.)
| | - Maria J. Garcia-Velloso
- Department of Nuclear Medicine, University Clinic of Navarra, Center of Applied Medical Research (CIMA), Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain; (M.I.M.-L.); (E.P.)
- Correspondence: ; Tel.: +34-948-255400 (ext. 4948); Fax: +34-948-296500
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, 97080 Würzburg, Germany; (O.V.); (S.S.); (A.K.B.); (C.L.)
- Nuclear Medicine, Medical Faculty, University of Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| |
Collapse
|
38
|
Abstract
Hybrid imaging using the tracer [18F]FDG (2‑deoxy-2-fluoro-D-glucose) is regarded as the backbone of the diagnostic workup of lymphomas. All international guidelines, and especially the Lugano and RECIL (Response Evaluation Criteria in Lymphoma) guidelines, currently recommend [18F]FDG-PET/CT (positron emission tomography/computed tomography) for staging and treatment response assessment. With the exception of pediatric lymphomas, neither PET/MRI (magnetic resonance imaging) nor whole-body MRI are currently endorsed by international guidelines, despite the fact that both techniques have clear advantages over [18F]FDG-PET/CT in the assessment of lymphomas with variable FDG avidity. Of the new, more specific PET tracers that are being evaluated for the use in lymphomas, the CXCR4 (CXC motif chemokine receptor 4) tracer [68Ga]Pentixafor is of particular interest, as initial studies have shown that it may be used to visualize frequently non-FDG-avid lymphomas such as small-cell lymphocytic lymphoma, mucosa-associated lymphoid tissue (MALT) lymphoma and lymphomplasmacytic lymphoma.
Collapse
Affiliation(s)
- Marius E Mayerhöfer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, 10065, New York, USA.
- Univ.-Klinik für Radiologie und Nuklearmedizin, Abteilung für Allgemeine und Kinderradiologie, Medizinische Universität Wien, Währinger Gürtel 18-20, Wien, 1090, Österreich.
| | - Alexander Haug
- Univ.-Klinik für Radiologie und Nuklearmedizin, Abteilung für Nuklearmedizin, Medizinische Universität Wien, Währinger Gürtel 18-20, Wien, 1090, Österreich
| |
Collapse
|
39
|
Methionine Dependence of Cancer. Biomolecules 2020; 10:biom10040568. [PMID: 32276408 PMCID: PMC7226524 DOI: 10.3390/biom10040568] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/25/2022] Open
Abstract
Tumorigenesis is accompanied by the reprogramming of cellular metabolism. The shift from oxidative phosphorylation to predominantly glycolytic pathways to support rapid growth is well known and is often referred to as the Warburg effect. However, other metabolic changes and acquired needs that distinguish cancer cells from normal cells have also been discovered. The dependence of cancer cells on exogenous methionine is one of them and is known as methionine dependence or the Hoffman effect. This phenomenon describes the inability of cancer cells to proliferate when methionine is replaced with its metabolic precursor, homocysteine, while proliferation of non-tumor cells is unaffected by these conditions. Surprisingly, cancer cells can readily synthesize methionine from homocysteine, so their dependency on exogenous methionine reflects a general need for altered metabolic flux through pathways linked to methionine. In this review, an overview of the field will be provided and recent discoveries will be discussed.
Collapse
|
40
|
Abstract
This review discusses nuclear imaging of inflammation using molecular probes beyond fluoro-d-glucose, is structured by cellular targets, and focuses on those tracers that have been successfully applied clinically.
Collapse
Affiliation(s)
- Malte Kircher
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany.
| |
Collapse
|
41
|
Oliva S, D'Agostino M, Boccadoro M, Larocca A. Clinical Applications and Future Directions of Minimal Residual Disease Testing in Multiple Myeloma. Front Oncol 2020; 10:1. [PMID: 32076595 PMCID: PMC7006453 DOI: 10.3389/fonc.2020.00001] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
In the last years, the life expectancy of multiple myeloma (MM) patients has substantially improved thanks to the availability of many new drugs. Our ability to induce deep responses has improved as well, and the treatment goal in patients tolerating treatment moved from the delay of progression to the induction of the deepest possible response. As a result of these advances, a great scientific effort has been made to redefine response monitoring, resulting in the development and validation of high-sensitivity techniques to detect minimal residual disease (MRD). In 2016, the International Myeloma Working Group (IMWG) updated MM response categories defining MRD-negative responses both in the bone marrow (assessed by next-generation flow cytometry or next-generation sequencing) and outside the bone marrow. MRD is an important factor independently predicting prognosis during MM treatment. Moreover, using novel combination therapies, MRD-negative status can be achieved in a fairly high percentage of patients. However, many questions regarding the clinical use of MRD status remain unanswered. MRD monitoring can guide treatment intensity, although well-designed clinical trials are needed to demonstrate this potential. This mini-review will focus on currently available techniques and data on MRD testing and their potential future applications.
Collapse
Affiliation(s)
- Stefania Oliva
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Mattia D'Agostino
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Alessandra Larocca
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| |
Collapse
|
42
|
Sachpekidis C, Goldschmidt H, Dimitrakopoulou-Strauss A. Positron Emission Tomography (PET) Radiopharmaceuticals in Multiple Myeloma. Molecules 2019; 25:molecules25010134. [PMID: 31905752 PMCID: PMC6982887 DOI: 10.3390/molecules25010134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 12/24/2019] [Accepted: 12/26/2019] [Indexed: 12/31/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell disorder, characterized by clonal proliferation of malignant plasma cells in the bone marrow. Bone disease is the most frequent feature and an end-organ defining indicator of MM. In this context, imaging plays a pivotal role in the management of the malignancy. For several decades whole-body X-ray survey (WBXR) has been applied for the diagnosis and staging of bone disease in MM. However, the serious drawbacks of WBXR have led to its gradual replacement from novel imaging modalities, such as computed tomography (CT), magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT). PET/CT, with the tracer 18F-fluorodeoxyglucose (18F-FDG), is now considered a powerful diagnostic tool for the detection of medullary and extramedullary disease at the time of diagnosis, a reliable predictor of survival as well as the most robust modality for treatment response evaluation in MM. On the other hand, 18F-FDG carries its own limitations as a radiopharmaceutical, including a rather poor sensitivity for the detection of diffuse bone marrow infiltration, a relatively low specificity, and the lack of widely applied, established criteria for image interpretation. This has led to the development of several alternative PET tracers, some of which with promising results regarding MM detection. The aim of this review article is to outline the major applications of PET/CT with different radiopharmaceuticals in the clinical practice of MM.
Collapse
Affiliation(s)
- Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center, 69120 Heidelberg, Germany;
- Correspondence: or ; Tel.: +49-6221-42-2478; Fax: +49-6221-42-2476
| | - Hartmut Goldschmidt
- Department of Internal Medicine V, University Hospital Heidelberg and National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany;
| | | |
Collapse
|
43
|
Da Vià MC, Solimando AG, Garitano-Trojaola A, Barrio S, Munawar U, Strifler S, Haertle L, Rhodes N, Teufel E, Vogt C, Lapa C, Beilhack A, Rasche L, Einsele H, Kortüm KM. CIC Mutation as a Molecular Mechanism of Acquired Resistance to Combined BRAF-MEK Inhibition in Extramedullary Multiple Myeloma with Central Nervous System Involvement. Oncologist 2019; 25:112-118. [PMID: 32043788 DOI: 10.1634/theoncologist.2019-0356] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 08/20/2019] [Indexed: 12/21/2022] Open
Abstract
Combined MEK-BRAF inhibition is a well-established treatment strategy in BRAF-mutated cancer, most prominently in malignant melanoma with durable responses being achieved through this targeted therapy. However, a subset of patients face primary unresponsiveness despite presence of the activating mutation at position V600E, and others acquire resistance under treatment. Underlying resistance mechanisms are largely unknown, and diagnostic tests to predict tumor response to BRAF-MEK inhibitor treatment are unavailable. Multiple myeloma represents the second most common hematologic malignancy, and point mutations in BRAF are detectable in about 10% of patients. Targeted inhibition has been successfully applied, with mixed responses observed in a substantial subset of patients mirroring the widespread spatial heterogeneity in this genomically complex disease. Central nervous system (CNS) involvement is an extremely rare, extramedullary form of multiple myeloma that can be diagnosed in less than 1% of patients. It is considered an ultimate high-risk feature, associated with unfavorable cytogenetics, and, even with intense treatment applied, survival is short, reaching less than 12 months in most cases. Here we not only describe the first patient with an extramedullary CNS relapse responding to targeted dabrafenib and trametinib treatment, we furthermore provide evidence that a point mutation within the capicua transcriptional repressor (CIC) gene mediated the acquired resistance in this patient. KEY POINTS: BRAF mutations constitute an attractive druggable target in multiple myeloma. This is the first genomic dissection of the central nervous system involvement in a multiple myeloma patient harboring a druggable BRAFV600E mutation. Deep genomic characterization of the extramedullary lesion prompted a personalized therapeutic approach. Acquisition of CIC mutation confers a mechanism of BRAF-MEK inhibitor drug resistance in multiple myeloma. The in silico interrogation of the CoMMpass clinical study revealed 10 patients with somatic mutations of CIC and its downregulation at gene expression level in multiple myeloma. CIC gene silencing decreases the sensitivity of multiple myeloma cells to BRAF-MEK inhibition in vitro. The correlation between CIC downregulation and ETV4/5 nuclear factor expression in multiple myeloma BRAF-mutant cells is shown for the first time. CIC mutation, its downregulation, and the related downstream effect on MMP24 support disseminative potential providing new clues in the extramedullary biology definition.
Collapse
Affiliation(s)
| | - Antonio Giovanni Solimando
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy
| | | | - Santiago Barrio
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Umair Munawar
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Susanne Strifler
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Larissa Haertle
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Nadine Rhodes
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Eva Teufel
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Cornelia Vogt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
44
|
Abstract
PURPOSE PET/CT with both C-choline and C-methionine has recently been reported to offer advantages over F-FDG for imaging in multiple myeloma (MM). The aim of this study was to directly compare the diagnostic performance of both non-FDG radiotracers in MM patients. METHODS Nineteen patients with a history of MM (n = 18) or solitary bone plasmacytoma (n = 1) underwent both C-choline and C-methionine PET/CT for diagnostic imaging. In this retrospective analysis, scans were compared on a patient and on a lesion basis. In 12 patients, respective tracer uptake in the iliac crest was correlated with the extent of malignant bone marrow plasma cell infiltration. RESULTS C-methionine detected more intramedullary MM lesions in 8 (42.1%) of 19 patients. In the remainder (11/19 [57.9%]), both C-choline and C-methionine provided equal results. C-methionine demonstrated higher lesion-to-muscle ratios (P = 0.0001). In the 12 patients in whom a recent bone marrow biopsy was available, SUVmean as well as SUVmax correlated significantly with the degree of malignant plasma cell infiltration for both C-methionine (SUVmean: r = 0.85, P < 0.001; SUVmax: r = 0.82, P = 0.001) and C-choline (SUVmean: r = 0.72, P < 0.008; SUVmax: r = 0.73; P = 0.006). CONCLUSIONS Our data suggest that C-methionine PET/CT might be more sensitive than C-choline PET/CT for the detection of active MM lesions.
Collapse
|
45
|
Solovev MV, Mendeleeva LP, Firsova MV, Aslanidi IP, Mukhortova OV, Savchenko VG. [PET/CT with 18F-fluorodeoxyglucose and 11C-methionine after autologous stem cell transplantation in multiple myeloma patients]. TERAPEVT ARKH 2019; 91:75-82. [PMID: 32598739 DOI: 10.26442/00403660.2019.07.000328] [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: 04/16/2020] [Indexed: 11/22/2022]
Abstract
AIM to compare the results of tumor visualization when using 18F-FDG and 11C-methionine PET/CT after auto-HSCT in MM patients. MATERIALS AND METHODS A prospective study included 27 MM patients subjected to 18F-FDG and 11C-methionine PET/CT on day 100 after auto-HSCT. Obtained images were visually and semi - quantitatively analyzed. Focal areas of increased uptake for every radiopharmaceutical agent (hypermetabolic foci) not associated with its physiological distribution were registered. Maximum Standardized Uptake Values (SUVmax) in pathological foci were automatically calculated for every radiopharmaceutical agent separately. PET/CT findings were compared to antitumor response achieved after auto-HSCT according to International MM Working Group criteria. RESULTS After auto-HSCT, the majority of patients (16/60%) achieved a complete response. Abnormal 18F-FDG uptake was registered in 37% (n=10) of patients, negative PET findings were obtained in 63% (n=17) of patients. 11C-methionine PET/CT revealed hypermetabolic foci in 67% (n=18) of patients, and there was no 11C-methionine uptake in 33% (n=9). Pathological foci of radiopharmaceutical agent uptake were 1.8 times more frequently revealed using PET/CT with 11C-methionine (p.
Collapse
Affiliation(s)
| | | | | | - I P Aslanidi
- Bakoulev Scientific Center for Cardiovascular Surgery
| | | | | |
Collapse
|
46
|
Jamet B, Bailly C, Carlier T, Touzeau C, Nanni C, Zamagni E, Barré L, Michaud AV, Chérel M, Moreau P, Bodet-Milin C, Kraeber-Bodéré F. Interest of Pet Imaging in Multiple Myeloma. Front Med (Lausanne) 2019; 6:69. [PMID: 31024917 PMCID: PMC6465522 DOI: 10.3389/fmed.2019.00069] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/20/2019] [Indexed: 12/15/2022] Open
Abstract
The interest of 18Fluoro-deoxyglucose (FDG) positron emission tomography (PET) imaging in the management of patients with multiple myeloma (MM) for the workup at diagnosis and for therapeutic evaluation has recently been demonstrated. FDG-PET is a powerful imaging tool for bone lesions detection at initial diagnosis with high sensitivity and specificity values. The independent pejorative prognostic value on progression-free survival (PFS) and overall survival (OS) of baseline PET-derived parameters (presence of extra-medullary disease (EMD), number of focal bone lesions (FLs), and maximum standardized uptake values [SUVmax]) has been reported in several large independent prospective studies. During therapeutic evaluation, FDG-PET is considered as the reference imaging technique, because it can be performed much earlier than MRI which lacks specificity. Persistence of significant FDG uptake after treatment, notably before maintenance therapy, is an independent pejorative prognostic factor, especially for patients with a complete biological response. So FDG-PET and medullary flow cytometry are complementary tools for detection of minimal residual disease before maintenance therapy. However, the definition of PET metabolic complete response should be standardized. In patients with smoldering multiple myeloma, the presence of at least one hyper-metabolic lytic lesions on FDG-PET may be considered as a criterion for initiating therapy. FDG-PET is also indicated for initial staging of a solitary plasmacytoma so as to not disregard other bone or extra-medullary localizations. Development of nuclear medicine offer new perspectives for MM imaging. Recent PET tracers are willing to overcome limitations of FDG. (11)C-Methionine, which uptake reflects the increased protein synthesis of malignant cells seems to correlate well with bone marrow infiltration. Lipid tracers, such as Choline or acetate, and some peptide tracers, such as (68) Ga-Pentixafor, that targets CXCR4 (chemokine receptor-4, which is often expressed with high density by myeloma cells), are other promising PET ligands. 18F-fludarabine and immuno-PET targeting CD138 and CD38 also showed promising results in preclinical models.
Collapse
Affiliation(s)
- Bastien Jamet
- Nuclear Medicine Unit, University Hospital, Nantes, France
| | - Clément Bailly
- Nuclear Medicine Unit, University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Nantes University, Nantes, France
| | - Thomas Carlier
- Nuclear Medicine Unit, University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Nantes University, Nantes, France
| | - Cyrille Touzeau
- CRCINA, INSERM, CNRS, Nantes University, Nantes, France.,Haematology Department, University Hospital, Nantes, France
| | - Cristina Nanni
- Nuclear Medicine, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elena Zamagni
- Seràgnoli Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | | | | | - Michel Chérel
- CRCINA, INSERM, CNRS, Nantes University, Nantes, France
| | - Philippe Moreau
- CRCINA, INSERM, CNRS, Nantes University, Nantes, France.,Haematology Department, University Hospital, Nantes, France
| | - Caroline Bodet-Milin
- Nuclear Medicine Unit, University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Nantes University, Nantes, France
| | - Françoise Kraeber-Bodéré
- Nuclear Medicine Unit, University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Nantes University, Nantes, France.,Nuclear Medicine Unit, ICO-Gauducheau, Nantes-Saint-Herblain, France
| |
Collapse
|
47
|
Withofs N, Beguin Y, Cousin F, Tancredi T, Simoni P, Alvarez-Miezentseva V, De Prijck B, Hafraoui K, Bonnet C, Baron F, Hustinx R, Caers J. Dual-tracer PET/CT scan after injection of combined [ 18 F]NaF and [ 18 F]FDG outperforms MRI in the detection of myeloma lesions. Hematol Oncol 2019; 37:193-201. [PMID: 30821017 DOI: 10.1002/hon.2600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/05/2019] [Accepted: 02/26/2019] [Indexed: 12/16/2022]
Abstract
The detection rates of whole-body combined [18 F]NaF/[18 F]FDG positron emission tomography combined with computed tomography (PET/CT), CT alone, whole-body magnetic resonance imaging (WB-MRI), and X-ray were prospectively studied in patients with treatment-requiring plasma cell disorders The detection rates of imaging techniques were compared, and focal lesions were classified according to their anatomic location. Twenty-six out of 30 initially included patients were assessable. The number of focal lesions detected in newly diagnosed patients (n = 13) and in relapsed patients (n = 13) were 296 and 234, respectively. The detection rate of PET/CT was significantly higher than those of WB-MRI (P < 0.05) and CT (P < 0.0001) both in patients with newly diagnosed and in those with relapsed multiple myeloma (MM). The X-ray detection rate was significantly lower than those of all other techniques, while CT detected more lesions compared with WB-MRI at diagnosis (P = 0.025). With regard to the infiltration patters, relapsed patients presented more diffuse patterns, and more focal lesions located in the limbs compared with newly diagnosed patients. In conclusion, the detection rate of [18 F]NaF/[18 F]FDG PET/CT was significantly higher than those of CT, MRI, and X-ray, while the detection rate of X-rays was significantly lower than those of all other imaging techniques except for focal lesions located in the skull.
Collapse
Affiliation(s)
- Nadia Withofs
- Division of Nuclear Medicine and Oncological Imaging, Medical Physics Department, CHU of Liège, Liège, Belgium
| | - Yves Beguin
- Department of Clinical Hematology, CHU of Liège, Liège, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - François Cousin
- Division of Nuclear Medicine and Oncological Imaging, Medical Physics Department, CHU of Liège, Liège, Belgium
| | - Tino Tancredi
- Division of Nuclear Medicine and Oncological Imaging, Medical Physics Department, CHU of Liège, Liège, Belgium
| | - Paolo Simoni
- Division of Diagnostic Imaging, Medical Physics Department, CHU of Liège, Liège, Belgium
| | | | | | | | | | - Frédéric Baron
- Department of Clinical Hematology, CHU of Liège, Liège, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, Medical Physics Department, CHU of Liège, Liège, Belgium
| | - Jo Caers
- Department of Clinical Hematology, CHU of Liège, Liège, Belgium.,Laboratory of Hematology, GIGA-I3, University of Liège, Liège, Belgium
| |
Collapse
|
48
|
Abstract
Bone disease is the most frequent feature of multiple myeloma (MM) and represents a marker of end-organ damage; it is used to establish the diagnosis and to dictate the immediate need for therapy. For this reason, imaging plays a significant role in the management of MM patients. Although conventional radiography has traditionally been the standard imaging modality, its low sensitivity in detecting osteolytic lesions and inability to evaluate response to therapy has called for the use of more sophisticated techniques, such as whole-body low-dose computed tomography (WBLDCT), whole-body magnetic resonance imaging, and 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (PET/CT). In this review, the advantages, indications of use, and applications of the 3 techniques in the management of patients with MM in different settings will be discussed. The European Myeloma Network and the European Society for Medical Oncology guidelines have recommended WBLDCT as the imaging modality of choice for the initial assessment of MM-related lytic bone lesions. Magnetic resonance imaging is the gold-standard imaging modality for detection of bone marrow involvement, whereas PET/CT provides valuable prognostic data and is the preferred technique for assessment of response to therapy. Standardization of most of the techniques is ongoing.
Collapse
|
49
|
Rasche L, Alapat D, Kumar M, Gershner G, McDonald J, Wardell CP, Samant R, Van Hemert R, Epstein J, Williams AF, Thanendrarajan S, Schinke C, Bauer M, Ashby C, Tytarenko RG, van Rhee F, Walker BA, Zangari M, Barlogie B, Davies FE, Morgan GJ, Weinhold N. Combination of flow cytometry and functional imaging for monitoring of residual disease in myeloma. Leukemia 2018; 33:1713-1722. [PMID: 30573775 PMCID: PMC6586541 DOI: 10.1038/s41375-018-0329-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/24/2018] [Accepted: 10/10/2018] [Indexed: 02/08/2023]
Abstract
The iliac crest is the sampling site for minimal residual disease (MRD) monitoring in multiple myeloma (MM). However, the disease distribution is often heterogeneous, and imaging can be used to complement MRD detection at a single site. We have investigated patients in complete remission (CR) during first-line or salvage therapy for whom MRD flow cytometry and the two imaging modalities positron emission tomography (PET) and diffusion-weighted magnetic resonance imaging (DW-MRI) were performed at the onset of CR. Residual focal lesions (FLs), detectable in 24% of first-line patients, were associated with short progression-free survival (PFS), with DW-MRI detecting disease in more patients. In some patients, FLs were only PET positive, indicating that the two approaches are complementary. Combining MRD and imaging improved prediction of outcome, with double-negative and double-positive features defining groups with excellent and dismal PFS, respectively. FLs were a rare event (12%) in first-line MRD-negative CR patients. In contrast, patients achieving an MRD-negative CR during salvage therapy frequently had FLs (50%). Multi-region sequencing and imaging in an MRD-negative patient showed persistence of spatially separated clones. In conclusion, we show that DW-MRI is a promising tool for monitoring residual disease that complements PET and should be combined with MRD.
Collapse
Affiliation(s)
- L Rasche
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Internal Medicine 2, University Hospital of Würzburg, Würzburg, Germany
| | - D Alapat
- Pathology Department, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - M Kumar
- Radiology Department, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - G Gershner
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - J McDonald
- Radiology Department, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - C P Wardell
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - R Samant
- Radiology Department, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - R Van Hemert
- Radiology Department, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - J Epstein
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - A F Williams
- Pathology Department, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - S Thanendrarajan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - C Schinke
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - M Bauer
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - C Ashby
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - R G Tytarenko
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - F van Rhee
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - B A Walker
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - M Zangari
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - B Barlogie
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - F E Davies
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - G J Morgan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - N Weinhold
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
50
|
Kircher S, Stolzenburg A, Kortüm KM, Kircher M, Da Via M, Samnick S, Buck AK, Einsele H, Rosenwald A, Lapa C. Hexokinase-2 Expression in 11C-Methionine–Positive, 18F-FDG–Negative Multiple Myeloma. J Nucl Med 2018; 60:348-352. [DOI: 10.2967/jnumed.118.217539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/10/2018] [Indexed: 12/13/2022] Open
|