Next Article in Journal
Prospects of Intraoperative Multimodal OCT Application in Patients with Acute Mesenteric Ischemia
Next Article in Special Issue
Magnetic Resonance Imaging for Surveillance of Hepatocellular Carcinoma: A Systematic Review and Meta-Analysis
Previous Article in Journal
Nourin-Dependent miR-137 and miR-106b: Novel Biomarkers for Early Diagnosis of Myocardial Ischemia in Coronary Artery Disease Patients
Previous Article in Special Issue
Concordance between Response Assessment Using Prostate-Specific Membrane Antigen PET and Serum Prostate-Specific Antigen Levels after Systemic Treatment in Patients with Metastatic Castration Resistant Prostate Cancer: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Comparison of [18F]FDG PET/CT and MRI for Treatment Response Assessment in Multiple Myeloma: A Meta-Analysis

Department of Diagnostic Radiology, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
*
Author to whom correspondence should be addressed.
Diagnostics 2021, 11(4), 706; https://doi.org/10.3390/diagnostics11040706
Submission received: 11 March 2021 / Revised: 10 April 2021 / Accepted: 13 April 2021 / Published: 15 April 2021

Abstract

:
The present study was designed to assess the additional value of 2-deoxy-2[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) to magnetic resonance imaging (MRI) in the treatment response assessment of multiple myeloma (MM). We performed a meta-analysis of all available studies to compare the detectability of treatment response of [18F]FDG PET/CT and MRI in treated MM. We defined detecting a good therapeutic effect as positive, and residual disease as negative. We determined the sensitivities and specificities across studies, calculated the positive and negative likelihood ratios (LR), and made summary receiver operating characteristic curves (SROC) using hierarchical regression models. The pooled analysis included six studies that comprised 278 patients. The respective performance characteristics (95% confidence interval (CI)) of [18F]FDG PET/CT and MRI were as follows: sensitivity of 80% (56% to 94%) and 25% (19% to 31%); specificity of 58% (44% to 71%) and 83% (71% to 91%); diagnostic odds ratio (DOR) of 6.0 (3.0–12.0) and 1.7 (0.7–2.7); positive LR of 1.8 (1.3–2.4) and 1.4 (0.7–2.7); and negative LR of 0.33 (0.21–0.53) and 0.81 (0.62–1.1). In the respective SROC curves, the area under the curve was 0.77 (SE, 0.038) and 0.59 (SE, 0.079) and the Q* index was 0.71 and 0.57. Compared with MRI, [18F]FDG PET/CT had higher sensitivity and better DOR and SROC curves. Compared with MRI, [18F]FDG PET/CT had greater ability to detect the treatment assessment of MM.

1. Introduction

Multiple myeloma (MM) is the second most common hematologic cancer, and frequently develops in the elderly, with 65 years as the median age at diagnosis. MM accounts for approximately 1% of cancers and 10% of all hematologic malignancies [1,2]. It is characterized by the proliferation of plasma cells that produce abnormal monoclonal immunoglobulin, which infiltrates the bone marrow (BM), and by the excessive production of monoclonal immunoglobulins that can be detected in serum and/or urine [1]. The major clinical manifestations of MM have been given the acronym CRAB, which comprise hypercalcemia (C) and bone destruction (B) secondary to overactivation of osteoclasts; renal impairment (R), which is mostly caused by monoclonal light chains that affect the kidneys, and anemia (A) [1]. BM involvement and extramedullary disease (EMD) have been recognized as important factors that influence the prognosis and clinical management of patients with MM [3,4].
Because 80% to 90% of all patients with MM develop BM involvement [5], imaging has an increasingly important role in the assessment of the degree of BM involvement and treatment effect. A conventional radiographic skeletal survey was historically used for the assessment of bone lesions in patients with MM. However, many studies have shown higher sensitivity for the detection of focal lesions with whole-body computed tomography (CT), 2-deoxy-2[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) and whole-body magnetic resonance imaging (MRI) than with conventional radiographic skeletal surveys. Among these, whole-body MRI is the most sensitive technique for the visualization of focal and diffuse infiltration of the BM in untreated patients [6,7]. Similarly, [18F]FDG PET/CT has been recognized as a sensitive tool to assess the extent of both BM involvement and EMD in newly diagnosed MM [8,9,10]. Both MRI and [18F]FDG PET/CT upon diagnosis and during follow-up have been shown to be of prognostic value for progression-free survival (PFS) and/or overall survival (OS) [7,11].
Studies that compared the diagnostic performance between whole-body MRI and [18F]FDG PET/CT for the initial pretreatment staging of MM suggested that MRI had higher sensitivity in lesion detection, allowing the detection of both diffuse BM infiltration and focal lesions before destruction of the mineral bone [6,12,13,14]. Although, the International Myeloma Working Group (IMWG) recommended whole-body CT or PET/CT for first-line imaging to detect osteolytic lesions, the Myeloma Response Assessment and Diagnosis System (MY-RADS) guidelines and the National Institute for Health and Care Excellence (NICE) guidelines in the UK recommend MRI as the first-line of imaging for all patients with a suspected diagnosis of asymptomatic myeloma or solitary bone plasmacytoma [5,15,16]. The role of [18F]FDG PET/CT remains limited in the initial assessment. However, in treatment assessment, [18F]FDG PET/CT, compared with MRI, was associated with faster normalization of imaging findings in patients who had achieved complete response (CR) or very good partial response (PR) after therapy [7,17,18,19]. Moreover, compared with MRI, [18F]FDG PET/CT showed better prognostic value in detecting EMD and minimal residual disease [5,9,11,20]. Current evidence shows that [18F]FDG-PET is a sensitive tool for the evaluation of early therapeutic response in MM. Many studies have shown the value of [18F]FDG PET/CT in the treatment response assessment in MM; however, the IMWG recommendation is still weak [5]. Therefore, strong evidence on the routine use of [18F]FDG PET/CT in clinical decision-making is required. The present study was designed to perform a meta-analysis of all available studies and to assess the advantage of additional [18F]FDG PET/CT over MRI in the treatment response assessment in MM.

2. Materials and Methods

2.1. Data Sources Eligibility

We searched PubMed/MEDLINE, SCOPUS, and Biological Abstracts from inception to January 2021. Search queries included terms related to “multiple myeloma,” “[18F]FDG PET,” “MRI,” and “treatment response”. Different keywords including Medical Subject Heading (MeSH) terms were combined using Boolean operations ‘AND’ and ‘OR’ such as (“PET” OR “Positron Emission Tomography”[MeSH]) AND (“FDG”[MeSH] OR “Fluoro-Deoxy-Glucose”) AND (“MRI”[MeSH] OR “Magnetic resonance imaging” OR “DWI” OR “diffusion weighted imaging” OR “ADC”) AND (“Multiple myeloma [MeSH]” OR “MM”) AND (“response” OR “surveillance” OR “treatment effect”). We did not apply any language restrictions. Two reviewers (KY and JT) independently assessed the potentially relevant citations for inclusion, and disagreements were resolved by consensus. The referenced articles of the retrieved articles were screened to identify additional relevant articles.
The inclusion criteria were made based upon the Patient/Intervention/Comparator/Outcome/Study design (PICOS) criteria [21] as follows: (1) “patients” with MM that were given treatment, such as chemotherapy and/or stem cell transplantation (SCT), (2) [18F]FDG PET or PET/CT for the treatment response assessment or surveillance after treatment as the “intervention,” (3) MRI as the “comparator”, (4) detection of treatment response with IMWG Uniform Response Criteria for Multiple Myeloma as “outcomes,” and (5) the “study design” included prospective or retrospective studies published as original articles or brief communications. The exclusion criteria were as follows: (1) small sample size (<10 patients), (2) were not in the field of interest, (3) had an overlap in the study population with another report, or (4) had insufficient information for derivation of 2 × 2 tables to draw a summary receiver operating characteristic curve (SROC). Reviews, guidelines, consensus statement, editorial, letters, book chapter, case reports, and meeting abstracts were also excluded.

2.2. Data Extraction

We independently extracted data from the eligible studies and resolved any issues by consensus. We recorded the author names; journal; publication year; origin country; number of patients; age; inclusion and exclusion criteria; study design; treatment details; imaging details, such as imaging system (PET or PET/CT; 1.5T or 3T MRI) and protocols; number of experts who interpreted the images; and the qualitative or quantitative definition of a positive test result. The remission or relapsed status in the reference standard was recorded. In addition, the numbers of patients who showed treatment effects as good responders and poor responders on [18F]FDG PET/CT and MRI were extracted; 2 × 2 tables were created and included the numbers of true negative (TP), false positive (FP), false negative (FN), and true negative (TN). We defined detecting a good therapeutic effect as positive, and residual disease as negative. Data extracted from publications alone were deemed adequate for this meta-analysis without contacting the authors for more information. Two reviewers (KY and JT) individually applied the widely used Quality Assessment of Diagnostic Accuracy Studies-2 tool to assess the quality of the systematic reviews of diagnostic studies [22].

2.3. Statistical Analysis

To evaluate the diagnostic value of [18F]FDG PET/CT in the treatment response assessment of MM, we calculated the pooled estimates of sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratios (DORs), and their 95% confidence intervals (CI) for both, in [18F]FDG PET/CT and MRI. The calculation of LRs combined sensitivity and specificity. The PLR was defined as the ratio of sensitivity over (1-specificity), and the NLR was defined as the ratio of (1-sensitivity) over specificity. The DOR was calculated as the ratio of PLR relative to NLR, with higher values indicating better performance. In addition, SROCs were drawn, and area under the curve (AUC) and Q* index were obtained. The Q* index is the best statistical method to reflect diagnostic value and was defined by the point where the sensitivity and specificity were equal; it represented the point that was closest to the ideal top left corner of the SROC space [23]. The degree of heterogeneity among the different studies was tested using the I2 test. When significant heterogeneity (i.e., I2 value of >50%) was observed, a random effect model was applied; in the other cases, a fixed effect model was used [24]. Analyses were performed using Meta-Disc v. 1.4 (XI Cochrane Colloquium, Barcelona, Spain) and Review Manager (version 5.3; Cochrane Collaboration).

3. Results

3.1. Article Search

The initial electronic search retrieved 363 articles (Figure 1). Of these, 21 articles were retrieved with the corresponding full texts after removing duplicate and nonrelevant articles. Six original articles, including a total of 278 patients, were finally included in the quantitative synthesis [7,13,17,18,19,25]. The quality assessment of the six included studies is shown in Figure 2. A total of 278 patients with MM were analyzed for the diagnostic accuracy of treatment effect assessment of the imaging methods. All 278 patients were evaluated with MRI, and 272 of them were evaluated with additional [18F]FDG PET/CT.
Of the six eligible studies, four were prospective and two were retrospective. All the studies enrolled patients with MM and had undergone [18F]FDG PET/CT and MRI for treatment assessment. One study used 3T MRI and three studies used 1.5T MRI; two studies had no available details. In three studies, the whole-body was scanned through coronal images, whereas the other three studies acquired whole spine and pelvic images. Four studies used contrast medium and two studies used noncontrast images only. All studies used [18F]FDG PET/CT. The amounts of radiotracer injected during [18F]FDG PET/CT were 350, 360 and 370 Mbq, and 3–7 Mbq/kg and 5.3 Mbq/kg, respectively. The time interval was 60–80 minutes in all studies. The results of all studies were evaluated qualitatively. The reference standard comprised BM biopsy, laboratory data, MRI, and [18F]FDG PET/CT.Most of the studies followed the international uniform response criteria for MM [26,27]. The detailed characteristics of included studies are summarized in Table 1.

3.2. Pooled Diagnostic Performance (Meta-Analysis)

Figure 3 and Figure 4 show the combined SROC curves and their corresponding findings for MRI and [18F]FDG PET/CT, respectively. The findings of imaging modality are described below.
-
[18F]FDG PET or PET/CT
This pooled analysis included six studies that comprised 273 patients. The performance characteristics (95% CI) were sensitivity, 80% (56% to 94%); specificity, 58% (44% to 71%); DOR, 6.0 (3.0–12.0); PLR, 1.8 (1.3–2.4); and NLR, 0.33 (0.21–0.53). In the SROC curves, the AUC was 0.77 (SE, 0.038), and the Q* index was 0.71.
-
MRI
This pooled analysis included six studies that comprised 278 patients. The performance characteristics (95% CI) were sensitivity, 25% (19% to 31%); specificity, 83% (71% to 91%); DOR, 1.7 (0.7–4.5); PLR, 1.4 (0.7–2.7); and NLR, 0.81 (0.62–1.1). In the SROC curves, the AUC was 0.59 (SE, 0.079), and the Q* index was 0.57.
-
Comparison of imaging modalities
In summary, compared with MRI, [18F]FDG PET/CT had higher sensitivity (80%) and DOR (6.0). The SROC curves of the [18F]FDG PET/CT studies showed excellent diagnostic performance (Figure 3 and Figure 4). No publication bias was observed based on the results of Funnel plot analysis (Supplementary Materials).
For each test, we combined the summary receiver operating characteristic curves and their corresponding findings. FN, false negative; FP, false positive; TN, true negative; TP, true positive.

4. Discussion

Compared with MRI, [18F]FDG PET/CT had excellent detectability of treatment response in MM with higher sensitivity. Overall, the value of [18F]FDG PET/CT for the detection of early treatment response was higher compared with that of MRI. We believe that the main reason for the low sensitivity of MRI was the inability to distinguish between scar tissue and active disease after treatment. Previous studies on [18F]FDG PET/CT after treatment have shown a significant association between early metabolic response and histopathologic tumor regression [28,29]. In addition to its ability to detect early treatment response, [18F]FDG PET/CT showed a good prognostic value in MM. Completely negative [18F]FDG uptake after treatment was proven to correlate with a high-quality response to therapy in MM [10,11]. In two studies, [18F]FDG PET/CT was evaluated either on day seven after induction treatment or before the first autologous SCT. Persistence of >3 focal lesions on day seven was an early predictor of significantly shorter PFS and OS, especially in the subgroup of patients with high-risk gene expression profiles [30]. Although the value of [18F]FDG PET/CT in the assessment of treatment response has been well established, the recommendation for its routine use in clinical settings remains weak [5,31]. To our knowledge, this was the first meta-analysis that compared the diagnostic ability between [18F]FDG PET/CT and MRI in the assessment of treatment response in MM. Our results show evidence for the clinical use of [18F]FDG PET/CT in MM.
Based on the results of this meta-analysis, the performance of [18F]FDG PET/CT in the assessment of treatment effect was satisfactory, based on the high percentage of true positives and the low percentage of false negative results. On the other hand, the specificity was lower with [18F]FDG PET/CT than with MRI. A possible reason for this was the relatively high detection rate of transient or extremely fast response in patients who were yet to be assessed to have PR or very good PR according to the response criteria. The fact that [18F]FDG PET/CT showed good prognostic value might support the idea, because patients who have false positive results (i.e., persistent abnormal plasma cells on BM biopsy with positive treatment response on [18F]FDG PET/CT, are expected to have a high probability of good prognosis according to studies [11,19]. Therefore, patients who show good response on [18F]FDG PET/CT should be carefully followed-up and can be expected to achieve a good remission state. In follow-up, IMWG response criteria determine [18F]FDG uptake as positive or negative, depending on whether it is higher or lower than blood or surrounding normal tissue. However, in clinical practice, the diagnosis may be equivocal, as bone marrow also exhibits mild [18F]FDG uptake in response to normal metabolic or hematopoietic activity, and degenerative uptake may be seen in older generations. In the future, a scoring system is expected to be constructed that can evaluate more objectively in combination with CT and MRI.
In the present study, the MRI protocols mainly used conventional T1 weighted image (T1WI), T2 weighted image (T2WI), short TI inversion recovery (STIR), and contrast enhanced T1WI. However, a good diagnostic value of whole-body diffusion-weighted image (WBDWI) or PET/MRI has been reported in the treatment assessment of patients with MM [30,32,33,34,35]. The Myeloma Response Assessment and Diagnosis System (or MY-RADS) [16], a standardized evaluation method proposed in 2019, also recommends WBDWI according to its usefulness in previous reports [32,36,37,38]. These studies claim that WBDWI can detect more lesions, but it has not been confirmed whether the lesions detected are true lesions. The possibility of post-treatment changes or hyperplastic bone marrow has not been ruled out [39,40,41]. One study reported that [18F]FDG PET/CT was superior in detecting extramedullary lesions in treatment response assessment [42]. The fact that only intramedullary lesions are more detected in WBDWI indicates that it may also include benign changes such as hyperplastic bone marrow. In most studies, WBDWI detects more focal lesions than FDG PET/CT in newly diagnosed MM, but on a per-patient basis WBDWI and [18F]FDG PET-CT showed no differences in the detection of bone involvement, with almost perfect agreement between the two methods [43]. In addition, [18F]FDG PET-CT could be considered a high-performance imaging procedure for diagnosis of diffuse infiltration [43]. Nowadays, many studies on WBDWI have focused on the detection of BM lesions in newly diagnosed MM [40,44], and there are few studies assessed the correlation with IMWG response criteria in post-treatment evaluation [38]. A study that was not included in the quantitative evaluation of the present meta-analysis, due to the small number of cases, compared the ability of WBDWI and FDG PET/CT to evaluate treatment response in eight cases of MM. According to this, the sensitivity, specificity, and diagnostic accuracy were 60%, 33% and 50% for PET, and 20%, 67%, and 37.5% for MRI, respectively [38]. Although the number of cases was small and not statistically significant, [18F]FDG PET/CT appeared to be superior to WBDWI in assessing treatment response. Further validation studies that compare [18F]FDG PET/CT with WBDWI or PET/MRI are warranted.
The present study had certain limitations. First, the diagnostic value of [18F]FDG PET/CT in patients with MM was reported without adjusting for potential confounders, such as the grade and stage of MM. Moreover, this meta-analysis included a source of heterogeneity (i.e., publication bias), which may result in systematic differences in the derived effect size estimates from small and large studies. The available data from the enrolled studies consistently showed great improvement in sensitivity with [18F]FDG PET/CT over MRI. Although the confidence intervals included a low sensitivity for MRI and a low specificity for [18F]FDG PET/CT, the number of studies was limited to only those that had available data for deriving 2 × 2 tables to draw SROC. There was a risk for subjective interpretation because the [18F]FDG PET/CT in all the studies was qualitatively assessed by two reviewers; in addition, the presence/absence of blinding was unclear. Although the cost and radiation exposure of each procedure are important concerns for both the patient and the healthcare system, we were unable to analyze these because of the limited number of published articles. Selection of the most appropriate imaging test in any clinical situation would depend on the circumstances of the patient, expertise, available equipment at the treating site and procedure costs. Certainly, the goal is to use the most cost-effective imaging method that would allow accurate diagnosis and prompt treatment of this common, complex and costly condition. This will require further prospective studies on a larger sample size, with a direct comparison among the different radiologic and nuclear medicine techniques.
Nevertheless, our study provided evidence to support the possible application of [18F]FDG PET/CT, in addition to MRI, for the assessment of treatment response in patients with MM. Compared with MRI, [18F]FDG PET/CT had higher sensitivity for the detection of early treatment response. Further research is required to confirm the incremental diagnostic improvement with [18F]FDG PET/CT over MRI, with consideration of radiation dose, cost-effectiveness and the potential complications against the yield of information.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/diagnostics11040706/s1, Figure S1: Funnel plots for publication bias evaluation.

Author Contributions

U.T. conceived the study, and K.Y. and J.T. participated in data collection, and performed the analysis. All authors participated in drafting the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article or Supplementary Material.

Acknowledgments

The authors thank the Guideline Committee of the Japan Radiological Society (JRS) for the valuable assistance in the acquisition of articles and for the helpful suggestions. This study was supported in part by the National Cancer Center Research and Development Fund (2020-J-3). This study was also supported by the Ministry of Health, Labour and Welfare Grants and the Japan Agency for Medical Research and Development Grants No.15ck0106162h0001, 16ck0106162h0002, 20ck0106577h0001, and 17ck0106162h0003. We were given helpful suggestions by Daniel C Sullivan; Alexander Guimaraes, who is the Chair of the Steering Committee of the Quantitative Imaging Biomarkers Alliance (QIBA); the Radiological Society of North America; Shigeki Aoki; and Ukihide Tateishi, who is the Chair of the Steering Committee, J-QIBA, JRS.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rajkumar, S.V.; Dimopoulos, M.A.; Palumbo, A.; Blade, J.; Merlini, G.; Mateos, M.-V.; Kumar, S.; Hillengass, J.; Kastritis, E.; Richardson, P.; et al. International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma. Lancet Oncol. 2014, 15, e538–e548. [Google Scholar] [CrossRef]
  2. Rajkumar, S.V. Multiple myeloma: 2014 Update on diagnosis, risk-stratification, and management. Am. J. Hematol. 2014, 89, 998–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sager, S.; Ergül, N.; Ciftci, H.; Cetin, G.; Güner, S.I.; Cermik, T.F. The value of FDG PET/CT in the initial staging and bone marrow involvement of patients with multiple myeloma. Skelet. Radiol. 2011, 40, 843–847. [Google Scholar] [CrossRef] [PubMed]
  4. Zaidi, H.; Montandon, M.-L.; Alavi, A. The Clinical Role of Fusion Imaging Using PET, CT, and MR Imaging. Magn. Reson. Imaging Clin. N. Am. 2010, 18, 133–149. [Google Scholar] [CrossRef] [Green Version]
  5. Hillengass, J.; Usmani, S.; Rajkumar, S.V.; Durie, B.G.M.; Mateos, M.-V.; Lonial, S.; Joao, C.; Anderson, K.C.; García-Sanz, R.; Riva, E.; et al. International myeloma working group consensus recommendations on imaging in monoclonal plasma cell disorders. Lancet Oncol. 2019, 20, e302–e312. [Google Scholar] [CrossRef]
  6. Fonti, R.; Salvatore, B.; Quarantelli, M.; Sirignano, C.; Segreto, S.; Petruzziello, F.; Catalano, L.; Liuzzi, R.; Rotoli, B.; Del Vecchio, S.; et al. 18F-FDG PET/CT, 99mTc-MIBI, and MRI in Evaluation of Patients with Multiple Myeloma. J. Nucl. Med. 2008, 49, 195–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Derlin, T.; Peldschus, K.; Münster, S.; Bannas, P.; Herrmann, J.; Stübig, T.; Habermann, C.R.; Adam, G.; Kröger, N.; Weber, C. Comparative diagnostic performance of 18F-FDG PET/CT versus whole-body MRI for determination of remission status in multiple myeloma after stem cell transplantation. Eur. Radiol. 2012, 23, 570–578. [Google Scholar] [CrossRef] [PubMed]
  8. Durie, B.G.M.; Waxman, A.D.; D’Agnolo, A.; Williams, C.M. Whole-body (18)F-FDG PET identifies high-risk myeloma. J. Nucl. Med. 2002, 43. [Google Scholar]
  9. Nanni, C.; Zamagni, E.; Farsad, M.; Castellucci, P.; Tosi, P.; Cangini, D.; Salizzoni, E.; Canini, R.; Cavo, M.; Fanti, S. Role of 18F-FDG PET/CT in the assessment of bone involvement in newly diagnosed multiple myeloma: Preliminary results. Eur. J. Nucl. Med. Mol. Imaging 2006, 33, 525–531. [Google Scholar] [CrossRef] [PubMed]
  10. Bartel, T.B.; Haessler, J.; Brown, T.L.Y.; Shaughnessy, J.J.D.; Van Rhee, F.; Anaissie, E.; Alpe, T.; Angtuaco, E.; Walker, R.; Epstein, J.; et al. F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma. Blood 2009, 114, 2068–2076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Zamagni, E.; Patriarca, F.; Nanni, C.; Zannetti, B.; Englaro, E.; Pezzi, A.; Tacchetti, P.; Buttignol, S.; Perrone, G.; Brioli, A.; et al. Prognostic relevance of 18-F FDG PET/CT in newly diagnosed multiple myeloma patients treated with up-front autologous transplantation. Blood 2011, 118, 5989–5995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Hillengass, J.; Landgren, O. Challenges and opportunities of novel imaging techniques in monoclonal plasma cell disorders: Imaging “early myeloma”. Leuk. Lymphoma 2013, 54, 1355–1363. [Google Scholar] [CrossRef] [PubMed]
  13. Zamagni, E.; Nanni, C.; Patriarca, F.; Englaro, E.; Castellucci, P.; Geatti, O.; Tosi, P.; Tacchetti, P.; Cangini, D.; Perrone, G.; et al. A prospective comparison of 18F-fluorodeoxyglucose positron emission tomography-computed tomography, magnetic resonance imaging and whole-body planar radiographs in the assessment of bone disease in newly diagnosed multiple myeloma. Haematologica 2007, 92, 50–55. [Google Scholar] [CrossRef] [PubMed]
  14. Gariani, J.; Westerland, O.; Natas, S.; Verma, H.; Cook, G.; Goh, V. Comparison of whole body magnetic resonance imaging (WBMRI) to whole body computed tomography (WBCT) or 18 F-fluorodeoxyglucose positron emission tomography/CT (18 F-FDG PET/CT) in patients with myeloma: Systematic review of diagnostic performance. Crit. Rev. Oncol. 2018, 124, 66–72. [Google Scholar] [CrossRef] [Green Version]
  15. The National Institute for Health and Care Excellence. Myeloma: Diagnosis and Management 2016. Available online: https://www.nice.org.uk/guidance/ng35 (accessed on 8 December 2020).
  16. Messiou, C.; Hillengass, J.; Delorme, S.; Lecouvet, F.E.; Moulopoulos, L.A.; Collins, D.J.; Blackledge, M.D.; Abildgaard, N.; Østergaard, B.; Schlemmer, H.-P.; et al. Guidelines for Acquisition, Interpretation, and Reporting of Whole-Body MRI in Myeloma: Myeloma Response Assessment and Diagnosis System (MY-RADS). Radiology 2019, 291, 5–13. [Google Scholar] [CrossRef] [Green Version]
  17. Spinnato, P.; Bazzocchi, A.; Brioli, A.; Nanni, C.; Zamagni, E.; Albisinni, U.; Cavo, M.; Fanti, S.; Battista, G.; Salizzoni, E. Contrast enhanced MRI and 18F-FDG PET-CT in the assessment of multiple myeloma: A comparison of results in different phases of the disease. Eur. J. Radiol. 2012, 81, 4013–4018. [Google Scholar] [CrossRef]
  18. Basha, M.A.A.; Hamed, M.A.G.; Refaat, R.; AlAzzazy, M.Z.; Bessar, M.A.; Mohamed, E.M.; Ahmed, A.F.; Tantawy, H.F.; Altaher, K.M.; Obaya, A.A.; et al. Diagnostic performance of 18F-FDG PET/CT and whole-body MRI before and early after treatment of multiple myeloma: A prospective comparative study. Jpn. J. Radiol. 2018, 36, 382–393. [Google Scholar] [CrossRef]
  19. Moreau, P.; Attal, M.; Caillot, D.; Macro, M.; Karlin, L.; Garderet, L.; Facon, T.; Benboubker, L.; Escoffre-Barbe, M.; Stoppa, A.-M.; et al. Prospective Evaluation of Magnetic Resonance Imaging and [18F]Fluorodeoxyglucose Positron Emission Tomography-Computed Tomography at Diagnosis and Before Maintenance Therapy in Symptomatic Patients With Multiple Myeloma Included in the IFM/DFCI 2009 Trial: Results of the IMAJEM Study. J. Clin. Oncol. 2017, 35, 2911–2918. [Google Scholar] [CrossRef]
  20. Lu, Y.-Y.; Chen, J.-H.; Lin, W.-Y.; Liang, J.-A.; Wang, H.-Y.; Tsai, S.-C.; Kao, C.-H. FDG PET or PET/CT for Detecting Intramedullary and Extramedullary Lesions in Multiple Myeloma. Clin. Nucl. Med. 2012, 37, 833–837. [Google Scholar] [CrossRef]
  21. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G.; For the PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. BMJ 2009, 339, b2535. [Google Scholar] [CrossRef] [Green Version]
  22. Whiting, P.F.; Rutjes, A.W.; Westwood, M.E.; Mallett, S.; Deeks, J.J.; Reitsma, J.B.; Leeflang, M.M.; Sterne, J.A.; Bossuyt, P.M.M.; The QUADAS-2 Group. QUADAS-2: A Revised Tool for the Quality Assessment of Diagnostic Accuracy Studies. Ann. Intern. Med. 2011, 155, 529–536. [Google Scholar] [CrossRef]
  23. Zamora, J.; Abraira, V.; Muriel, A.; Khan, K.; Coomarasamy, A. Meta-DiSc: A software for meta-analysis of test accuracy data. BMC Med. Res. Methodol. 2006, 6, 31. [Google Scholar] [CrossRef] [PubMed]
  24. Higgins, J.P.T.; Thompson, S.G. Quantifying heterogeneity in a meta-analysis. Stat. Med. 2002, 21, 1539–1558. [Google Scholar] [CrossRef] [PubMed]
  25. Cascini, G.L.; Falcone, C.; Console, D.; Restuccia, A.; Rossi, M.; Parlati, A.; Tamburrini, O. Whole-body MRI and PET/CT in multiple myeloma patients during staging and after treatment: Personal experience in a longitudinal study. La Radiol. Med. 2013, 118, 930–948. [Google Scholar] [CrossRef] [PubMed]
  26. Bladé, J.; Samson, D.; Reece, D.; Apperley, J.; Björkstrand, B.; Gahrton, G.; Gertz, M.; Giralt, S.; Jagannath, S.; Vesole, D. Criteria for Evaluating Disease Response and Progression in Patients with Multiple Myeloma Treated by High-Dose Therapy and Haemopoietic Stem Cell Transplantation. Br. J. Haematol. 1998, 102, 1115–1123. [Google Scholar] [CrossRef]
  27. Durie, B.G.M.; Harousseau, J.-L.; Miguel, J.S.; Bladé, J.; Barlogie, B.; Anderson, K.; Gertz, M.; Dimopoulos, M.; Westin, J.; Sonneveld, P.; et al. International uniform response criteria for multiple myeloma. Leukemia 2006, 20, 1467–1473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Gallamini, A.; Zwarthoed, C.; Borra, A. Positron Emission Tomography (PET) in Oncology. Cancers 2014, 6, 1821–1889. [Google Scholar] [CrossRef] [Green Version]
  29. Ben-Haim, S.; Ell, P. 18F-FDG PET and PET/CT in the Evaluation of Cancer Treatment Response. J. Nucl. Med. 2008, 50, 88–99. [Google Scholar] [CrossRef] [Green Version]
  30. Usmani, S.Z.; Mitchell, A.; Waheed, S.; Crowley, J.; Hoering, A.; Petty, N.; Brown, T.; Bartel, T.; Anaissie, E.; Van Rhee, F.; et al. Prognostic implications of serial 18-fluoro-deoxyglucose emission tomography in multiple myeloma treated with total therapy. Blood 2013, 121, 1819–1823. [Google Scholar] [CrossRef] [Green Version]
  31. Caers, J.; Garderet, L.; Kortüm, K.M.; O’Dwyer, M.E.; Van De Donk, N.W.; Binder, M.; Dold, S.M.; Gay, F.; Corre, J.; Beguin, Y.; et al. European Myeloma Network recommendations on tools for the diagnosis and monitoring of multiple myeloma: What to use and when. Haematologica 2018, 103, 1772–1784. [Google Scholar] [CrossRef]
  32. Pawlyn, C.; Fowkes, L.; Otero, S.; Jones, J.R.; Boyd, K.D.; Davies, F.E.; Morgan, G.J.; Collins, D.J.; Sharma, B.; Riddell, A.; et al. Whole-body diffusion-weighted MRI: A new gold standard for assessing disease burden in patients with multiple myeloma? Leukemia 2016, 30, 1446–1448. [Google Scholar] [CrossRef] [Green Version]
  33. Horger, M.; Weisel, K.; Horger, W.; Mroue, A.; Fenchel, M.; Lichy, M. Whole-Body Diffusion-Weighted MRI With Apparent Diffusion Coefficient Mapping for Early Response Monitoring in Multiple Myeloma: Preliminary Results. Am. J. Roentgenol. 2011, 196, W790–W795. [Google Scholar] [CrossRef]
  34. Mulé, S.; Reizine, E.; Blanc-Durand, P.; Baranes, L.; Zerbib, P.; Burns, R.; Nouri, R.; Itti, E.; Luciani, A. Whole-Body Functional MRI and PET/MRI in Multiple Myeloma. Cancers 2020, 12, 3155. [Google Scholar] [CrossRef] [PubMed]
  35. Sachpekidis, C.; Hillengass, J.; Goldschmidt, H.; Mosebach, J.; Pan, L.; Schlemmer, H.-P.; Haberkorn, U.; Dimitrakopoulou-Strauss, A. Comparison of 18F-FDG PET/CT and PET/MRI in patients with multiple myeloma. Am. J. Nucl. Med. Mol. Imaging 2015, 5, 469–478. [Google Scholar]
  36. Messiou, C.; Kaiser, M. Whole body diffusion weighted MRI—A new view of myeloma. Br. J. Haematol. 2015, 171, 29–37. [Google Scholar] [CrossRef]
  37. Pearce, T.; Philip, S.; Brown, J.; Koh, D.M.; Burn, P.R. Bone metastases from prostate, breast and multiple myeloma: Differences in lesion conspicuity at short-tau inversion recovery and diffusion-weighted MRI. Br. J. Radiol. 2012, 85, 1102–1106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Sachpekidis, C.; Mosebach, J.; Freitag, M.T.; Wilhelm, T.; Mai, E.K.; Goldschmidt, H.; Haberkorn, U.; Schlemmer, H.-P.; Delorme, S.; Dimitrakopoulou-Strauss, A. Application of 18F-FDG PET and diffusion weighted imaging (DWI) in multiple myeloma: Comparison of functional imaging modalities. Am. J. Nucl. Med. Mol. Imaging 2015, 5, 479–492. [Google Scholar] [PubMed]
  39. León, N.G.; Bueno, B.A.; Pérez, M.H.; Ramírez, L.F.L.; Alegre, A.; Colletti, P.M.; Rubello, D.; Carreras, J.L.; Bolton, R.C.D. Agreement Between 18F-FDG PET/CT and Whole-Body Magnetic Resonance Compared with Skeletal Survey for Initial Staging and Response at End-of-Treatment Evaluation of Patients with Multiple Myeloma. Clin. Nucl. Med. 2021, 46, 310–322. [Google Scholar] [CrossRef]
  40. Westerland, O.; Amlani, A.; Kelly-Morland, C.; Fraczek, M.; Bailey, K.; Gleeson, M.; El-Najjar, I.; Streetly, M.; Bassett, P.; Cook, G.J.R.; et al. Comparison of the diagnostic performance and impact on management of 18F-FDG PET/CT and whole-body MRI in multiple myeloma. Eur. J. Nucl. Med. Mol. Imaging 2021, 1–8. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Xiong, X.; Fu, Z.; Dai, H.; Yao, F.; Liu, D.; Deng, S.; Hu, C. Whole-body diffusion-weighted MRI for evaluation of response in multiple myeloma patients following bortezomib-based therapy: A large single-center cohort study. Eur. J. Radiol. 2019, 120, 108695. [Google Scholar] [CrossRef]
  42. Chen, J.; Li, C.; Tian, Y.; Xiao, Q.; Deng, M.; Hu, H.; Wen, B.; He, Y. Comparison of Whole-Body DWI and 18F-FDG PET/CT for Detecting Intramedullary and Extramedullary Lesions in Multiple Myeloma. Am. J. Roentgenol. 2019, 213, 514–523. [Google Scholar] [CrossRef] [PubMed]
  43. Mesguich, C.; Hulin, C.; Latrabe, V.; Lascaux, A.; Bordenave, L.; Hindié, E.; Marit, G. Prospective comparison of 18-FDG PET/CT and whole-body diffusion-weighted MRI in the assessment of multiple myeloma. Ann. Hematol. 2020, 99, 2869–2880. [Google Scholar] [CrossRef] [PubMed]
  44. Takasu, M.; Kondo, S.; Akiyama, Y.; Takahashi, Y.; Maeda, S.; Baba, Y.; Kawase, T.; Ichinohe, T.; Awai, K. Assessment of early treatment response on MRI in multiple myeloma: Comparative study of whole-body diffusion-weighted and lumbar spinal MRI. PLoS ONE 2020, 15, e0229607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. PRISMA flow diagram of included articles. PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses.
Figure 1. PRISMA flow diagram of included articles. PRISMA, Preferred Reporting Items for Systematic Reviews and Meta-Analyses.
Diagnostics 11 00706 g001
Figure 2. Quality assessment of the included studies using QUADAS-2.
Figure 2. Quality assessment of the included studies using QUADAS-2.
Diagnostics 11 00706 g002
Figure 3. Forest plots of the sensitivity and specificity and the pooled diagnostic performance of [18F]FDG PET/CT and MRI.
Figure 3. Forest plots of the sensitivity and specificity and the pooled diagnostic performance of [18F]FDG PET/CT and MRI.
Diagnostics 11 00706 g003
Figure 4. Summary receiver operating characteristic curves (SROC) for the diagnostic performance of [18F]FDG PET/CT and MRI. The size of the circle diamond indicates the weight of each study in [18F]FDG PET/CT (black curve) and MRI (red curve) respectively. The area under the SROC curve is 0.76 for [18F]FDG PET/CT and 0.59 for MRI.
Figure 4. Summary receiver operating characteristic curves (SROC) for the diagnostic performance of [18F]FDG PET/CT and MRI. The size of the circle diamond indicates the weight of each study in [18F]FDG PET/CT (black curve) and MRI (red curve) respectively. The area under the SROC curve is 0.76 for [18F]FDG PET/CT and 0.59 for MRI.
Diagnostics 11 00706 g004
Table 1. Study characteristics for selected studies.
Table 1. Study characteristics for selected studies.
AuthorYearRef.CountryNMean Age (Range)DesignTreatment ProtocolModalityFDG DoseTime IntervalFasting TimeNo. of AssessorsReference StandardDuration
Cascini GL, et al. 2013 [25] Italy22NR (44–83)PC or SCTPET/CT, 1.5T MRI360 MBq60 min 6 h NRIMWG criteria [27]2006.7–2010.3
Derlin T, et al. 2013 [7] Germany3155 (38–73)RSCTPET/CT, 1.5T MRI350 MBq60 min 4 h 2IMWG criteria [27]2008.7–2009.3
Elena Z, et al. 2007 [13]Italy 22 55 (42–65) PSCTPET/CT, MRI5.3 MBq/kg60–80 min6 h 2 EBMT criteria [26]2003.3–2004.12
Mohammad A, et al. 2018 [18] Egypt2262 (41–78)P18: C, 3: Ra, 1: SCT PET/CT, 3T MRI370 MBq60 min 6 h 3BM aspiration and biopsy2015.8–2017.12
Philippe M, et al. 2017 [19] France11959 (37–65)PC or SCTPET/CT, MRI3-7 MBq/kg60–80 min4 h 2IMWG criteria [27]2010.11–2012.11
Spinnato P, et al. 2012 [17] Italy5562 (33–81)RC or SCTPET/CT, 1.5T MRINRNR NA 2biopsy, L/D, MRI, PET/CT2009–2012.3
N number of patients, NR not reported, P prospective, R retrospective, C chemotherapy, SCT stem cell transplantation, Ra radiotherapy, IMWG International Myeloma Working Group, EBMT European Group for Blood and Marrow Transplant, BM bone marrow, L/D laboratory data.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yokoyama, K.; Tsuchiya, J.; Tateishi, U. Comparison of [18F]FDG PET/CT and MRI for Treatment Response Assessment in Multiple Myeloma: A Meta-Analysis. Diagnostics 2021, 11, 706. https://doi.org/10.3390/diagnostics11040706

AMA Style

Yokoyama K, Tsuchiya J, Tateishi U. Comparison of [18F]FDG PET/CT and MRI for Treatment Response Assessment in Multiple Myeloma: A Meta-Analysis. Diagnostics. 2021; 11(4):706. https://doi.org/10.3390/diagnostics11040706

Chicago/Turabian Style

Yokoyama, Kota, Junichi Tsuchiya, and Ukihide Tateishi. 2021. "Comparison of [18F]FDG PET/CT and MRI for Treatment Response Assessment in Multiple Myeloma: A Meta-Analysis" Diagnostics 11, no. 4: 706. https://doi.org/10.3390/diagnostics11040706

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop