1
|
Sagar S, Khan D, Sivasankar KV, Kumar R. New PET Tracers for Symptomatic Myeloma. PET Clin 2024; 19:515-524. [PMID: 39025753 DOI: 10.1016/j.cpet.2024.06.001] [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] [Indexed: 07/20/2024]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by the clonal proliferation of plasma cells within the bone marrow. Accurate staging and monitoring of disease progression are crucial for effective management. PET imaging has emerged as a powerful tool in the diagnosis and management of MM, with radiotracers like 18F-fluorodeoxyglucose and novel agents playing a pivotal role. This review explores the current state of PET imaging in multiple myeloma, focusing on its role in initial staging, response assessment, and prognosis prediction, with an emphasis on recent advancements.
Collapse
Affiliation(s)
- Sambit Sagar
- Diagnostic Nuclear Medicine Division, Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | - Dikhra Khan
- Diagnostic Nuclear Medicine Division, Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India
| | | | - Rakesh Kumar
- Diagnostic Nuclear Medicine Division, Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, Delhi, India.
| |
Collapse
|
2
|
Marchiori S, Cousin F, Papadopoulos I, Bernard C, Thys M, De Prijck B, Pirotte M, Donneau AF, Hustinx R, Caers J, Withofs N. Prognostic value of visual IMPeTUs criteria and metabolic tumor burden at baseline [ 18F]FDG PET/CT in patients with newly diagnosed multiple myeloma. EJNMMI Res 2024; 14:51. [PMID: 38806885 PMCID: PMC11133264 DOI: 10.1186/s13550-024-01113-6] [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: 04/05/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG) positron emission tomography combined with low-dose computed tomography (PET/CT) can be used at diagnosis to identify myeloma-defining events and also provides prognostic factors. The aim of this study was to assess the prognostic significance of baseline [18F]FDG PET/CT visual IMPeTUs (Italian myeloma criteria for PET Use)-based parameters and/or total metabolic tumor volume (TMTV) in a single-center population of patients with newly diagnosed multiple myeloma (NDMM) eligible for transplantation. METHODS Patients with MM who underwent a baseline [18F]FDG PET/CT were retrospectively selected from a large internal database of the University Hospital of Liege (Liege, Belgium). Initially, all PET/CT images were visually analyzed using IMPeTUs criteria, followed by delineation of TMTV using a semi-automatic lesion delineation workflow, including [18F]FDG-positive MM focal lesions (FL) with an absolute SUV threshold set at 4.0. In a first step, to ensure PET/CT scans accurate reporting, the agreement between two nuclear medicine physicians with distinct experience was assessed. In the second step, univariable and multivariable analyses were conducted to determine the prognostic significance of [18F]FDG PET/CT parameters on progression free survival (PFS) and overall survival (OS), respectively. RESULTS A total of 40 patients with NDMM were included in the study. The observers agreement in the analysis [18F]FDG PET/CT images was substantial for the presence of spine FL, extra spine FL, at least one fracture and paramedullary disease (Cohen's kappa 0.79, 0.87, 0.75 and 0.64, respectively). For the presence of skull FL and extramedullary disease the agreement was moderate (Cohen's kappa 0.56 and 0.53, respectively). Among [18F]FDG PET/CT parameters, a high number of delineated volumes of interest (VOI) using the SUV4.0 threshold was the only independent prognostic factor associated with PFS [HR (95% CI): 1.03 (1.004-1.05), P = 0.019] while a high number of FL (n > 10; F group 4) was the only independent prognostic factor associated with OS [HR (95% CI): 19.10 (1.90-191.95), P = 0.01]. CONCLUSION Our work confirms the reproducibility IMPeTUs criteria. Furthermore, it demonstrates that a high number of FL (n > 10; IMPeTUs F group 4), reflecting a high [18F]FDG-avid tumor burden, is an independent prognostic factor for OS. The prognostic value of the TMTV delineated using a SUV4.0 threshold was not significant. Nevertheless, the count of delineated [18F]FDG-avid lesions VOI using a SUV4.0 threshold was an independent prognostic factor for PFS.
Collapse
Affiliation(s)
- Silvano Marchiori
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liège, Belgium.
| | - François Cousin
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liège, Belgium
| | - Iraklis Papadopoulos
- Biostatistics and Research Methods Center (B-STAT), University of Liege, Liège, Belgium
| | - Claire Bernard
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liège, Belgium
| | - Marie Thys
- Medico-Economic Information Department / Data Analysis, CHU of Liège, Liège, Belgium
| | | | | | | | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liège, Belgium
- GIGA-CRC in Vivo Imaging, University of Liege, Liège, Belgium
| | - Jo Caers
- Department of Hematology, CHU de Liège, Liège, Belgium
- Laboratory of Hematology, GIGA I3, University of Liège, Liège, Belgium
| | - Nadia Withofs
- Division of Nuclear Medicine and Oncological Imaging, Department of Medical Physics, CHU of Liege, Liège, Belgium
- GIGA-CRC in Vivo Imaging, University of Liege, Liège, Belgium
| |
Collapse
|
3
|
Wadhwa A, Wang S, Patiño-Escobar B, Bidkar AP, Bobba KN, Chan E, Meher N, Bidlingmaier S, Su Y, Dhrona S, Geng H, Sarin V, VanBrocklin HF, Wilson DM, He J, Zhang L, Steri V, Wong SW, Martin TG, Seo Y, Liu B, Wiita AP, Flavell RR. CD46-Targeted Theranostics for PET and 225Ac-Radiopharmaceutical Therapy of Multiple Myeloma. Clin Cancer Res 2024; 30:1009-1021. [PMID: 38109209 PMCID: PMC10905524 DOI: 10.1158/1078-0432.ccr-23-2130] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/26/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023]
Abstract
PURPOSE Multiple myeloma is a plasma cell malignancy with an unmet clinical need for improved imaging methods and therapeutics. Recently, we identified CD46 as an overexpressed therapeutic target in multiple myeloma and developed the antibody YS5, which targets a cancer-specific epitope on this protein. We further developed the CD46-targeting PET probe [89Zr]Zr-DFO-YS5 for imaging and [225Ac]Ac-DOTA-YS5 for radiopharmaceutical therapy of prostate cancer. These prior studies suggested the feasibility of the CD46 antigen as a theranostic target in multiple myeloma. Herein, we validate [89Zr]Zr-DFO-YS5 for immunoPET imaging and [225Ac]Ac-DOTA-YS5 for radiopharmaceutical therapy of multiple myeloma in murine models. EXPERIMENTAL DESIGN In vitro saturation binding was performed using the CD46 expressing MM.1S multiple myeloma cell line. ImmunoPET imaging using [89Zr]Zr-DFO-YS5 was performed in immunodeficient (NSG) mice bearing subcutaneous and systemic multiple myeloma xenografts. For radioligand therapy, [225Ac]Ac-DOTA-YS5 was prepared, and both dose escalation and fractionated dose treatment studies were performed in mice bearing MM1.S-Luc systemic xenografts. Tumor burden was analyzed using BLI, and body weight and overall survival were recorded to assess antitumor effect and toxicity. RESULTS [89Zr]Zr-DFO-YS5 demonstrated high affinity for CD46 expressing MM.1S multiple myeloma cells (Kd = 16.3 nmol/L). In vitro assays in multiple myeloma cell lines demonstrated high binding, and bioinformatics analysis of human multiple myeloma samples revealed high CD46 expression. [89Zr]Zr-DFO-YS5 PET/CT specifically detected multiple myeloma lesions in a variety of models, with low uptake in controls, including CD46 knockout (KO) mice or multiple myeloma mice using a nontargeted antibody. In the MM.1S systemic model, localization of uptake on PET imaging correlated well with the luciferase expression from tumor cells. A treatment study using [225Ac]Ac-DOTA-YS5 in the MM.1S systemic model demonstrated a clear tumor volume and survival benefit in the treated groups. CONCLUSIONS Our study showed that the CD46-targeted probe [89Zr]Zr-DFO-YS5 can successfully image CD46-expressing multiple myeloma xenografts in murine models, and [225Ac]Ac-DOTA-YS5 can effectively inhibit the growth of multiple myeloma. These results demonstrate that CD46 is a promising theranostic target for multiple myeloma, with the potential for clinical translation.
Collapse
Affiliation(s)
- Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Bonell Patiño-Escobar
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Anil P. Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Emily Chan
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, California
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Vishesh Sarin
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - David M. Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia
| | - Li Zhang
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Medicine, Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Veronica Steri
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Sandy W. Wong
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Thomas G. Martin
- Department of Medicine, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Bin Liu
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Anesthesia, University of California, San Francisco, California
| | - Arun P. Wiita
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Laboratory Medicine, University of California, San Francisco, California
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
- Chan Zuckerberg Biohub, San Francisco, California
| | - Robert R. Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Garrastachu Zumarán P, García Megías I, Mangas Losada M, Mendoza Melero A, Villanueva Torres A, Boulvard Chollet X, Romero Robles L, Hernández Pérez PM, Ramírez Lasanta R, Delgado Bolton RC. Multitracer PET/CT with [ 18F]Fluorodeoxiglucose and [ 18F]Fluorocholine in the Initial Staging of Multiple Myeloma Patients Applying the IMPeTus Criteria: A Pilot Study. Diagnostics (Basel) 2023; 13:diagnostics13091570. [PMID: 37174961 PMCID: PMC10177931 DOI: 10.3390/diagnostics13091570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/16/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Initial staging of patients diagnosed with multiple myeloma (MM) can lead to negative results using conventional diagnostic imaging workup, including [18F]Fluorodesoxiglucose ([18F]FDG) PET/CT. The aim of this prospective pilot study was to evaluate the diagnostic efficacy of [18F]Fluorocholine ([18F]FCH) PET/CT in the initial staging of MM patients who were candidates for autologous bone marrow transplant. Materials and Methods: The inclusion criteria of our study were: (a) patients diagnosed with MM; (b) candidates for autologous bone marrow transplant (AT); and (c) studied with [18F]FCH PET/CT and [18F]FDG PET/CT for initial staging less than 4 weeks apart. Imaging analysis included the presence of: bone marrow infiltration, focal bone lesions, and para-medullary or extra-medullary disease, according to the proposed IMPeTus criteria. The analysis was performed per lesion, per patient, and per location. Results: The study population included ten patients. Globally, [18F]FCH PET/CT showed bone marrow uptake in all the patients and visualised 16 more focal lesions than [18F]FDG PET/CT. One patient presented a plasmacytoma, detected by both tracers. Extra-medullary and para-medullary disease was identified with different degrees of uptake by both tracers. In summary, [18F]FCH PET seemed to be superior to [18F]FDG PET/CT in detecting focal bone lesions. SUVmax values were slightly higher in [18F]FCH PET/CT than in [18F]FDG PET/CT. Conclusions: Taking into account the small study population, according to our results, [18F]FCH PET/CT could be a useful tool for staging MM patients.
Collapse
Affiliation(s)
- Puy Garrastachu Zumarán
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - Irene García Megías
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - María Mangas Losada
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - Alejandro Mendoza Melero
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - Amós Villanueva Torres
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - Xavier Boulvard Chollet
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - Leonardo Romero Robles
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | | | - Rafael Ramírez Lasanta
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| | - Roberto C Delgado Bolton
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro, 26006 Logroño, Spain
- Centre for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, 26006 Logroño, Spain
| |
Collapse
|
7
|
Wang Y, Yee A, Bernstein Z, O'Donnell E, Raje N, Mahmood U. Carbon-11-Labeled Methionine PET/CT in Patients With FDG-Occult Multiple Myeloma: A Prospective Pilot Study. AJR Am J Roentgenol 2023; 220:578-579. [PMID: 36382911 DOI: 10.2214/ajr.22.28560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In a pilot study (ClinicalTrials.gov NCT02646085), seven patients with treated multiple myeloma and negative FDG PET/CT underwent amino acid imaging with 11C-methionine PET/CT. In five participants, 11C-methionine PET/CT showed focal uptake corresponding with lytic lesions; two to 18 lesions were found (SUVmax, 2.8-6.4). Findings indicated a potential role for 11C-methionine PET/CT in detecting residual disease after negative FDG PET/CT, thereby guiding further treatment.
Collapse
Affiliation(s)
- Yingbing Wang
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Wht 4-427, Boston, MA 02114
| | - Andrew Yee
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Zachary Bernstein
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | | | - Noopur Raje
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Umar Mahmood
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Wht 4-427, Boston, MA 02114
| |
Collapse
|
8
|
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
|
9
|
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]
|
10
|
Application of Metabolic Reprogramming to Cancer Imaging and Diagnosis. Int J Mol Sci 2022; 23:ijms232415831. [PMID: 36555470 PMCID: PMC9782057 DOI: 10.3390/ijms232415831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cellular metabolism governs the signaling that supports physiological mechanisms and homeostasis in an individual, including neuronal transmission, wound healing, and circadian clock manipulation. Various factors have been linked to abnormal metabolic reprogramming, including gene mutations, epigenetic modifications, altered protein epitopes, and their involvement in the development of disease, including cancer. The presence of multiple distinct hallmarks and the resulting cellular reprogramming process have gradually revealed that these metabolism-related molecules may be able to be used to track or prevent the progression of cancer. Consequently, translational medicines have been developed using metabolic substrates, precursors, and other products depending on their biochemical mechanism of action. It is important to note that these metabolic analogs can also be used for imaging and therapeutic purposes in addition to competing for metabolic functions. In particular, due to their isotopic labeling, these compounds may also be used to localize and visualize tumor cells after uptake. In this review, the current development status, applicability, and limitations of compounds targeting metabolic reprogramming are described, as well as the imaging platforms that are most suitable for each compound and the types of cancer to which they are most appropriate.
Collapse
|
11
|
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
|
12
|
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
|
13
|
Böckle D, Tabares P, Zhou X, Schimanski S, Steinhardt MJ, Bittrich M, Seebacher E, Ulbrich M, Wilnit A, Metz C, Heidemeier A, Bley T, Werner R, Buck A, Einsele H, Kortüm KM, Beilhack A, Rasche L. Minimal residual disease and imaging-guided consolidation strategies in newly diagnosed and relapsed refractory multiple myeloma. Br J Haematol 2022; 198:515-522. [PMID: 35582835 DOI: 10.1111/bjh.18249] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/08/2022] [Accepted: 05/02/2022] [Indexed: 12/16/2022]
Abstract
Measurement of minimal residual disease (MRD) by next-generation flow cytometry (NGF) is an important tool to define deep responses in multiple myeloma (MM). However, little is known about the value of combining NGF with functional imaging and its role for MRD-based consolidation strategies in clinical routine. In the present study, we report our experience investigating these issues with 102 patients with newly diagnosed (n = 57) and relapsed/refractory MM (n = 45). Imaging was performed using either positron emission tomography or diffusion-weighted magnetic resonance imaging. In all, 45% of patients achieved MRD-negativity on both NGF and imaging (double-negativity), and 8% and 40% of patients were negative on either NGF or imaging respectively. Thus, in a minority of patients imaging was the only technique to detect residual disease. Imaging-positivity despite negativity on NGF was more common in heavily pretreated disease (four or more previous lines) compared to newly diagnosed MM (p < 0.01). Among the 29 patients undergoing MRD-triggered consolidation, 51% responded with MRD conversion and 21% with improved serological response. MRD-triggered consolidation led to superior progression-free survival (PFS) when compared to standard treatment (p = 0.04). In conclusion, we show that combining NGF with imaging is helpful particularly in patients with heavily pretreated MM, and that MRD-based consolidation could lead to improved PFS.
Collapse
Affiliation(s)
- David Böckle
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Paula Tabares
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Xiang Zhou
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Sven Schimanski
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Maximilian J Steinhardt
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Max Bittrich
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Elena Seebacher
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Maria Ulbrich
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Amy Wilnit
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Corona Metz
- Department of Radiology, Würzburg University Hospital, Würzburg, Germany
| | - Anke Heidemeier
- Department of Radiology, Würzburg University Hospital, Würzburg, Germany
| | - Thorsten Bley
- Department of Radiology, Würzburg University Hospital, Würzburg, Germany
| | - Rudolf Werner
- Department of Nuclear Medicine, Würzburg University Hospital, Würzburg, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, Würzburg University Hospital, Würzburg, Germany
| | - Hermann Einsele
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - K Martin Kortüm
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Andreas Beilhack
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Interdisciplinary Center for Clinical Research Laboratory, Würzburg University Hospital, Würzburg, Germany
| | - Leo Rasche
- Division of Oncology and Hematology, Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany.,Mildred Scheel Early Career Center, Würzburg University Hospital, Würzburg, Germany
| |
Collapse
|
14
|
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
|
15
|
van Dijken BRJ, Ankrah AO, Stormezand GN, Dierckx RAJO, Jan van Laar P, van der Hoorn A. Prognostic value of 11C-methionine volume-based PET parameters in IDH wild type glioblastoma. PLoS One 2022; 17:e0264387. [PMID: 35213602 PMCID: PMC8880430 DOI: 10.1371/journal.pone.0264387] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/09/2022] [Indexed: 11/18/2022] Open
Abstract
Purpose 11C-Methionine (11C-MET) PET prognostication of isocitrate dehydrogenase (IDH) wild type glioblastomas is inadequate as conventional parameters such as standardized uptake value (SUV) do not adequately reflect tumor heterogeneity. We retrospectively evaluated whether volume-based parameters such as metabolic tumor volume (MTV) and total lesion methionine metabolism (TLMM) outperformed SUV for survival correlation in patients with IDH wild type glioblastomas. Methods Thirteen IDH wild type glioblastoma patients underwent preoperative 11C-MET PET. Both SUV-based parameters and volume-based parameters were calculated for each lesion. Kaplan-Meier curves with log-rank testing and Cox regression analysis were used for correlation between PET parameters and overall survival. Results Median overall survival for the entire cohort was 393 days. MTV (HR 1.136, p = 0.007) and TLMM (HR 1.022, p = 0.030) were inversely correlated with overall survival. SUV-based 11C-MET PET parameters did not show a correlation with survival. In a paired analysis with other clinical parameters including age and radiotherapy dose, MTV and TLMM were found to be independent factors. Conclusions MTV and TLMM, and not SUV, significantly correlate with overall survival in patients with IDH wild type glioblastomas. The incorporation of volume-based 11C-MET PET parameters may lead to a better outcome prediction for this heterogeneous patient population.
Collapse
Affiliation(s)
- Bart R. J. van Dijken
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- * E-mail:
| | - Alfred O. Ankrah
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gilles N. Stormezand
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Rudi A. J. O. Dierckx
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Peter Jan van Laar
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Radiology, Zorggroep Twente, Almelo and Hengelo, the Netherlands
| | - Anouk van der Hoorn
- Department of Radiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
FDG-PET/CT, a Promising Exam for Detecting High-Risk Myeloma Patients? Cancers (Basel) 2020; 12:cancers12061384. [PMID: 32481533 PMCID: PMC7352681 DOI: 10.3390/cancers12061384] [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: 05/06/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma (MM) is a haematological neoplasm characterized by a clonal proliferation of malignant plasma cells in the bone marrow. MM is associated with high morbidity and mortality and variable survival, which can be very short for some patients but over 10 years for others. These differences in survival are explained by intra- and inter-tumoral heterogeneity and demonstrate the potential benefits of adapting the treatment course for high-risk patients with a poorer prognosis. Indeed, identification of these high-risk patients is necessary and is based on the identification of high-risk biomarkers including clinical variables, genomics and imaging results. Positron emission tomography combined with computed tomography using 18F-deoxyfluoroglucose (FDG-PET/CT) is a reliable technique for the initial staging of patients with symptomatic multiple myeloma (MM), and has been included in the IMWG (International Myeloma Working Group) recommendations in 2019. According to clinical studies, FDG-PET/CT characteristics could be used to define high-risk patients at initial diagnosis of symptomatic MM. The goal of this review is to demonstrate the prognostic value of FDG-PET in symptomatic MM patients, particularly in identifying high-risk patients, and thus, to best adapt therapeutic management in the future.
Collapse
|