Abstract

Bone disease represents a hallmark feature of multiple myeloma (MM), affecting nearly all patients during the disease course. Morphological imaging techniques play a crucial role in detecting bone disease, whereas functional ones are also fundamental for the differentiation of active from inactive disease and prognostic stratification. The International Myeloma Working Group (IMWG) currently recommends whole-body low-dose computed tomography (WBLDCT) as the first-choice imaging technique for the diagnosis of bone disease, whereas magnetic resonance imaging (MRI) is recommended in cases without further myeloma-defining events. However, 18F-fluorodeoxyglucose–positron emission tomography/CT (18F-FDG–PET/CT) currently represents the standard imaging technique, because it combines both morphological and functional data. Indeed, it allows detection of bone lesions (alternatively to WBLDCT), prognostic stratification, and monitoring of treatment response, being recommended by the IMWG for the assessment of imaging minimal residual disease. The IMPeTUs (Italian Myeloma criteria for PET Use) have proposed a visual descriptive assessment of 18F-FDG–PET/CT, with standardized definitions of metabolic responses. However, the use of further functional imaging techniques is being investigated, with diffusion-weighted (DW)–MRI being related to very promising results regarding both staging and response assessment, to the extent that myeloma response assessment and diagnosis system guidelines have recently proposed a standardization of acquisition, interpretation and reporting of this technique in MM, and the British guidelines consider DW-MRI an alternative to 18F-FDG–PET/CT. This review summarizes current knowledge on the use of functional imaging techniques in MM and their incorporation in recommendations/guidelines, and discusses potential future developments in this setting.

Multiple myeloma (MM) represents the second most common hematologic malignancy. It is caused by infiltration and proliferation of clonal plasma cells (PCs), primarily within the bone marrow (BM), and it is preceded by asymptomatic precursor stages (monoclonal gammopathy of undetermined significance and smoldering MM [SMM]).1 

Among clinical manifestations, myeloma-related bone disease (MBD) is the most common, affecting about 80% of patients with newly diagnosed MM (NDMM) and nearly all patients during disease course. MBD may cause bone pain and skeletal-related events, including pathological fractures, spinal cord compression, and need for radiotherapy/surgical procedures.2,3 Current diagnostic criteria by the International Myeloma Working Group (IMWG) define MBD as the presence of ≥1 osteolytic lesion (≥5 mm in size) on skeletal surveys, computed tomography (CT), or the CT component of 18F-fluorodeoxyglucose–positron emission tomography/CT (18F-FDG–PET/CT). Alternatively, another myeloma-defining event (MDE) is the presence of >1 focal lesion (FL), each ≥5 mm in size, by magnetic resonance imaging (MRI),4 as 2 studies in SMM have shown 70% progression rate to symptomatic MM at 2 years with a median time-to-progression of 13 to 15 months.5,6 Furthermore, disruption of cortical bone with growth of tumor masses arising from skeletal lesions may be causative of paraskeletal disease (PSD), affecting 6% to 34% of patients, with similar incidence at diagnosis and relapsed/refractory MM (RRMM). Conversely, hematogenous spread of malignant PCs to soft tissues may result in extramedullary disease (EMD), affecting 0.5% to 4.8% of patients with NDMM, and 3.4% to 14% of patients with RRMM.7,8 

The IMWG recommends whole-body low-dose CT (WBLDCT) as the first-choice/minimum requirement imaging technique, being more sensitive than conventional skeletal radiography in detecting osteolytic lesions9; with 18F-FDG–PET/CT as a potential alternative. In cases without CT-assessed osteolytic lesions and absence of other MDEs, whole-body MRI (WB-MRI), or spine and pelvis MRI (axial skeleton MRI [AS-MRI]), if WB-MRI is unavailable, is recommended to unveil FLs.10 Indeed, MRI showed the highest sensitivity in detecting early bone damage and diffuse BM involvement, differentiating pathological from osteoporotic fractures, and studying cord compression or other neurologic events.11-14 

Though conventional morphological imaging techniques represent the mainstay of MBD detection and are available worldwide, functional techniques are useful for prognostic stratification and are mandatory for treatment response evaluation, allowing noninvasive assessment of tissue composition, metabolism, blood flow, or other features related to disease activity. This is even more important in nonsecretory disease, as response cannot be evaluated by monitoring biochemical markers. Indeed, current IMWG guidelines recommend integration of conventional hematologic response with minimal residual disease (MRD) evaluation, both by next-generation sequencing/next-generation flow and imaging. Indeed, potential EMD, spatial heterogeneity, and patchy BM infiltration may hinder MRD assessment on BM, making imaging MRD fundamental. 18F-FDG–PET/CT is currently considered the standard recommended technique in this setting.10,15 However, a role for further techniques is emerging. Herein, we focus on functional imaging in MM, regarding diagnosis/staging, prognostic stratification, and response assessment.

PET/CT is a dual imaging technique combining functional and morphological assessment. The former is based on evaluation of a radiotracer uptake (with a spatial resolution of ∼5 mm), which allows to study metabolism and proliferation, thus becoming a surrogate marker of disease activity. The latter is due to the CT portion, providing the correct anatomical localization of metabolic foci, and allowing the identification of osteolytic lesions, fractures, PSD, and EMD.

The standard field of view (FOV) in MM usually includes at least skull, AS, upper limbs, and femurs. PET acquisition begins about 60 minutes after radiotracer infusion, and has a duration of 15 to 20 minutes. Last-generation scanners, however, are characterized by longer FOV, reaching significantly higher sensitivity and better spatial resolution. These tomographs require lower scanning time and inferior doses of radiotracer, decreasing radiation exposure. Notably, renal failure and metallic implants, representing contraindications to contrast agent-based techniques and MRI, respectively, are not impediments for PET/CT. The IMWG and the European Association of Nuclear Medicine have published specific guidelines aiming at standardizing acquisition, reporting and interpretation of 18F-FDG–PET/CT in PC dyscrasias.16,17 

Interpretation of PET/CT scans is based on both visual/qualitative and semiquantitative analyses. The latter uses a voxel-based parameter called standardized uptake value (SUV); specifically, SUVmax reflects the highest voxel value within the region of interest (ROI). Radiotracer uptake and SUV measurement are influenced by several factors, including patient’s weight and body composition, respiratory motion, tracer’s affinity, time elapsed from injection to image acquisition, location of the ROI, size of the matrix, and specific reconstruction parameters.18 As SUV refers to specific ROI, quantitative volumetric parameters have been introduced to assess the overall disease activity. The most commonly used are metabolic tumor volume (MTV) and total lesion glycolysis. However, their use in MM is currently limited by lack of standardization regarding delimitation of affected areas and definition of disease activity, to quantify tumor burden.19-23 

18F-FDG is the standardized most used radiotracer in MM, as it undergoes uptake, phosphorylation, and storage in metabolically active cells. False-negative scans may occur from recent high-dose steroid administration, hyperglycemia, or small skull lesions close to the brain. Furthermore, it was reported that ∼10% of patients have non–FDG-avid disease due to the lack of expression of glucose transporters or glycolysis enzymes, particularly hexokinase-2, potentially being not stable over time and changing during disease course. Conversely, false-positive FLs may result from artifacts (radiotracer accumulation in physiological districts, metallic implants), inflammation, infections, postsurgical areas or biopsies, fractures, or bone remodeling, whereas false diffuse disease (DD) may be caused by anemia or recent radiotherapy, chemotherapy, or growth factors administration.16,24 To overcome these limitations, several alternative tracers have been investigated, with some having potential theranostic implications. However, their use is limited by low availability and expertise, absence of standardization, need for local cyclotron for radiotracers with short half-life, and lack of prospective data.25,26 

The Italian Myeloma criteria for PET Use (IMPeTUs) have recently proposed a standardized assessment of 18F-FDG–PET/CT, based on visual descriptive integration of metabolic and anatomical characteristics. The formers are based on Deauville 5-point scale (DS), with active disease being DS ≥4 (Table 1).27 

Table 1.

IMPeTUs

Lesion typeSiteNumberGrading
Diffuse BM (“A” if hypermetabolism in limbs and ribs) — DS 
F (focal) S (skull) X1 (n = 0)  
 Sp (spine) X2 (n = 1-3)  
 Ex-Sp (extraspine) X3 (n = 4-10)  
  X4 (n > 10)  
L (lytic)   — 
Fr (fractures)  ≥1  
PSD   DS 
EMD N (nodal)    
 EN (extranodal)    
Lesion typeSiteNumberGrading
Diffuse BM (“A” if hypermetabolism in limbs and ribs) — DS 
F (focal) S (skull) X1 (n = 0)  
 Sp (spine) X2 (n = 1-3)  
 Ex-Sp (extraspine) X3 (n = 4-10)  
  X4 (n > 10)  
L (lytic)   — 
Fr (fractures)  ≥1  
PSD   DS 
EMD N (nodal)    
 EN (extranodal)    

Adapted from Nanni et al27 with permission.

For nodal N disease: C, cervical; SC, supraclavicular; M, mediastinal; Ax, axillary; Rp, retroperitoneal; Mes, mesentery; In, inguinal.

For extranodal disease: Li, liver; Mus, muscles; Spl, spleen; Sk, skin; Oth, other.

18F-FDG–PET/CT at staging

Current IMWG guidelines consider 18F-FDG–PET/CT as an alternative to WBLDCT regarding identification of osteolytic lesions as MDE, whereas radiotracer uptake without lytic lesions does not represent a criterion for treatment initiation.4 Its diagnostic role was supported by a study showing that risk of progression to active MM within 2 years for patients with SMM is 87% in cases with osteolytic lesions and 61% in cases of abnormal uptake without alterations in bone architecture, compared with 30% in patients with negative 18F-FDG–PET/CT.28 

Several studies regarding 18F-FDG–PET/CT in MM have reported 80% to 100% sensitivity and specificity in detecting bone lesions.12,29-32 Diagnostic performance of 18F-FDG–PET/CT has been evaluated by comparing several imaging techniques. Comparisons with whole-body skeletal radiography survey have demonstrated higher sensitivity of 18F-FDG–PET/CT in MBD diagnosis, except for skull and ribs lesions.12,14,31,33 A prospective comparison between the CT portion of 18F-FDG–PET/CT and WBLDCT revealed high concordance, suggesting the possibility to use the dual technique as the only method for MM evaluation.34 

Various studies also compared 18F-FDG–PET/CT with conventional MRI. Regarding DD, lower sensitivity of the former was generally observed. Conversely, FLs comparisons resulted differently based on the specific FOV of MRI, as 18F-FDG–PET/CT detects additional lesions to AS-MRI in ∼30% of patients, whereas diagnostic performance resulted similar or slightly inferior compared with WB-MRI.12,13,32,35,36 However, 18F-FDG–PET/CT has a greater impact on clinical decisions than WB-MRI.36 Conventional MRI was also found to detect earlier disease relapse than 18F-FDG–PET/CT.13 Comparisons to diffusion-weighted (DW)–MRI, described in the following paragraphs, showed inferior sensitivity in detecting FLs, PSD, and DD. Moreover, 18F-FDG–PET/CT resulted in 96% sensitivity and 78% specificity in detecting EMD, being therefore considered the best technique in this setting, except for central nervous system, for which MRI represents the mainstay (Figure 1).37 

Figure 1.

Disease patterns by 18F-FDG–PET/CT and WB-DW-MRI. (A) DD by 18F-FDG–PET/CT; (B) DD by WB-DW-MRI; (C) focal on DD by 18F-FDG–PET/CT; (D) focal on DD by WB-DW-MRI; (E) micronodular disease by WB-DW-MRI; (F) focal disease by 18F-FDG–PET/CT before (i) and after (ii) treatment; (G-H) focal disease by WB-DW-MRI before (i) and after (ii) treatment; (I) PSD by 18F-FDG–PET/CT (coronal, i; axial, ii); (J) PSD by WB-DW-MRI (coronal, i; axial, ii); (K) EMD by 18F-FDG–PET/CT (coronal, i; axial, ii); and (L) EMD by WB-DW-MRI (coronal, i; axial, ii).

Figure 1.

Disease patterns by 18F-FDG–PET/CT and WB-DW-MRI. (A) DD by 18F-FDG–PET/CT; (B) DD by WB-DW-MRI; (C) focal on DD by 18F-FDG–PET/CT; (D) focal on DD by WB-DW-MRI; (E) micronodular disease by WB-DW-MRI; (F) focal disease by 18F-FDG–PET/CT before (i) and after (ii) treatment; (G-H) focal disease by WB-DW-MRI before (i) and after (ii) treatment; (I) PSD by 18F-FDG–PET/CT (coronal, i; axial, ii); (J) PSD by WB-DW-MRI (coronal, i; axial, ii); (K) EMD by 18F-FDG–PET/CT (coronal, i; axial, ii); and (L) EMD by WB-DW-MRI (coronal, i; axial, ii).

Close modal

18F-FDG–PET/CT in prognostic stratification

Clinical outcomes of patients with MM are extremely heterogeneous, and prognostic stratification is essential to identify high-risk patients.

Several features assessed by 18F-FDG–PET/CT have proven prognostic in different settings. For example, focal FDG uptake without underlying osteolysis in patients with SMM was related to threefold risk of progression and shorter time-to-progression to active MM.38 In patients with NDMM, significant correlation of both progression-free survival (PFS) and overall survival (OS) with the presence of EMD, high number of FLs (>3) and elevated radiotracer uptake has been displayed by several studies and confirmed by recent meta-analyses.39,40 All these metabolic markers are related with specific biological aspects: EMD highlights metastatic properties with independence of clonal PCs from BM microenvironment and different disease biology; number of FLs is related to tumor burden; SUVmax to proliferation rate and aggressiveness. Notably, different values of SUVmax have been proposed: former experiences identified a cutoff value of ∼4 (mainly 4.2),29,41,42 whereas higher values (eg, >6.3 for FLs or >7.1 for BM diffuse uptake) were proposed in more recent trials.43 These characteristics could be integrated with conventional staging systems (International Staging System [ISS] and Revised ISS [R-ISS])42,44,45 or biological factors with established prognostic role (eg, circulating PCs)46 to improve patients’ stratification. As opposed to the presence of EMD, which has a well-known significant adverse outcome, discordant results have been produced regarding PSD, with some studies proposing it as an independent prognostic factor and others correlating outcomes with the number of PSD lesions.47-50 A prognostic role of volume parameters, including MTV, total lesion glycolysis, and the more recently described intensity of bone involvement and percentage of bone involvement has also been shown.20-23,51-53 

The prognostic role of 18F-FDG–PET/CT has also been assessed in RRMM: high number of FLs (with diverse cutoffs ranging from >3 to >10 in different studies), elevated SUVmax (with heterogeneous values, typically higher than NDMM setting), and presence of EMD have been related to patients’ outcome.54-58 Recent studies have also demonstrated a role of baseline 18F-FDG–PET/CT in the setting of novel immunotherapies: PET positivity, PSD, EMD, and high MTV before chimeric antigen receptor T-cell infusion have proven significantly prognostic.59-64 

Moreover, positivity of 18F-FDG–PET/CT resulted significantly associated to higher infiltration of BM-PCs, and the presence of high number of FLs to several markers of disease burden, including advanced staging according to Durie-Salmon system or elevated concentration of lactate dehydrogenase and β2-microglobulin.65 Furthermore, a significant link between high SUVmax and the presence of high-risk cytogenetic or molecular alterations, particularly del(17p)/TP53 mutation, has been observed.66 Overall, imaging characteristics maintain a significant independent prognostic role.52 

Notably, a prognostic role of disease characteristics assessed through IMPeTUs has been proposed: patients’ outcome has been related to the number of FLs and presence of DD, PSD, and EMD (NDMM); and presence of DD and EMD (RRMM).67-70 Therefore, an integration of IMPeTUs features to Durie-Salmon staging has been proposed (Durie-Salmon Plus), with good concordance to R-ISS.68 

18F-FDG–PET/CT in response assessment

The prognostic significance of 18F-FDG–PET/CT in response assessment has been demonstrated in different settings and at several time points. For example, early normalization of the metabolic scan in NDMM (7 days after induction initiation) is correlated with significantly improved PFS and OS, with a reversion of the adverse prognosis related to high number of baseline FLs, whereas persistence of >3 FLs is predictive of inferior outcome. Serial response assessment at different time points clarified that even later normalization and persistence of metabolic inactivity correlate with prognostic improvement, leading to conceptual treatment until suppression of FLs.41,71 

Furthermore, multiple studies have demonstrated a significant role of postinduction metabolic response, showing that normalization of 18F-FDG–PET/CT is significantly associated to improved PFS and OS. Conversely, persistence of active FLs and elevated uptake (eg, >4.2) before transplant procedures is predictive of worse prognosis (regardless of hematologic responses).29,30,32,41,42,72,73 “IMAJEM” highlighted that a SUVmax reduction >25% from baseline after induction therapy is related to significant PFS improvement.74 

However, the most important time point, currently recommended by IMWG guidelines, is before maintenance therapy.10 Indeed, several studies have demonstrated the fundamental prognostic role of metabolic response assessed 3 months after autologous stem cell transplantation (ASCT) or before maintenance, showing that normalization of metabolic activity is associated to longer PFS, OS, and time to relapse, and that residual SUVmax is inversely correlated to time to relapse.32,75,76 Moreover, the “CONPET” trial showed that consolidation treatment in patients with deep hematologic response but positive 18F-FDG–PET/CT after induction therapy and ASCT may lead to metabolic remission and likely to improved outcome.77 

The prognostic role of metabolic response has also been confirmed in RRMM. Indeed, a significant benefit was shown in patients achieving normalization of 18F-FDG–PET/CT after chimeric antigen receptor T-cell therapy, but data regarding the optimal timing for the assessment of response (1 or 3 months after infusion) are discordant.59,60,63 However, early evaluation of metabolic response in patients receiving T-cell engaging therapies has been associated to potential pseudoprogression due to flare-up phenomenon.78,79 

Noteworthy, the lack of standardization regarding definition of responses is a main limitation. Former studies defined disease activity based on heterogeneous SUVmax values.29,30 Nevertheless, because the radiotracer uptake may be influenced by several factors, a comparison to physiologic uptake in anatomic sites (DS) appears reasonable, and IMWG guidelines defined PET negativity as the disappearance of every area of increased tracer uptake, or a decrease to less than mediastinal blood pool SUV or less than surrounding normal tissue.15 However, a joint analysis of “EMN02” and “IFM2009,” based on evaluation of 18F-FDG–PET/CT according to IMPeTUs, has recently found that achievement of DS <4 (uptake lower than liver) for both FLs and BM diffuse uptake represents an independent predictor for improved PFS and OS. Therefore, new standardized definitions of metabolic responses have been proposed (Table 2).80 The applicability of these criteria was recently validated within the “FORTE” trial.81 

Table 2.

IMPeTUs and MY-RADS response criteria

Metabolic responseDescriptionRACsDescription
Complete metabolic response Uptake less than or equal to liver activity in BM sites and FLs previously involved, including PSD and EMD (DS 1-3) RAC1 Highly likely to be responding:
  • Return of normal fat containing marrow in areas previously infiltrated by FLs or DD;

  • Unequivocal decrease in number or size of FLs;

  • Conversion of a packed BM infiltrate into discrete nodules, with unequivocal decrease in tumor load in the respective BM space;

  • Decreasing soft tissue associated with bone disease;

  • Emergence of intra- or peritumoral fat within/around FLs (fat dot or halo sign);

  • Previously evident lesion shows increase in ADC from ≤1400 μm2/s to >1400 μm2/s;

  • ≥40% increase in ADC from baseline, with corresponding decrease in normalized high b-value signal intensity, and morphologic findings consistent with stable or responding disease;

  • For soft-tissue disease, RECIST version 1.1 criteria for CR (disappearance of all TLs)/PR (≥30% decrease in the sum of L-Di of TLs, taking as reference the baseline sum L-Di)

 
Partial metabolic response Decrease in number and/or activity of BM/FLs present at baseline, but persistence of lesion(s) with uptake more than liver activity (DS 4-5) RAC2 Likely to be responding:
  • Evidence of improvement but not enough to fulfill criteria for RAC1, for example, slight decrease in number/size of FLs, previously evident lesions showing increases in ADC from ≤1000 μm2/s to <1400 μm2/s, a 25% to 40% increase in ADC from baseline with corresponding decrease in b-value signal intensity, and morphologic findings consistent with stable or responding disease;

  • For soft-tissue disease, RECIST version 1.1 criteria not meeting requirements for PR

 
Stable metabolic disease No significant change in BM/FLs compared with baseline RAC3 No change:
  • No observable change

 
Progressive metabolic disease New FLs consistent with MM compared with baseline RAC4 Likely to be progressing:
  • Evidence of worsening disease, but not enough to fulfill criteria for RAC5;

  • Equivocal appearance of new lesion(s);

  • No change in size but increasing signal intensity on high b-value images (with ADC values <1400 μm2/s) consistent with possible disease progression;

  • Relapsed disease: reemergence of lesion(s) that previously disappeared or enlargement of lesion(s) that had partially regressed/stabilized with prior treatments;

  • Soft tissue in spinal canal causing narrowing not associated with neurologic findings and not requiring radiation therapy;

  • For soft-tissue disease, RECIST version 1.1 criteria not meeting requirements for PD

 
  RAC5 Highly likely to be progressing:
  • New critical fracture(s)/cord compression requiring radiation therapy/surgical intervention; only if confirmed as malignant with MRI signal characteristics;

  • Unequivocal new focal (5-10 mm)/diffuse area(s) of infiltration in regions of previously normal marrow;

  • Unequivocal increase in number/size of FLs;

  • Evolution of FLs to diffuse neoplastic pattern;

  • Appearance/increasing soft tissue associated with bone disease;

  • New lesions/regions of high signal intensity on high b-value images with ADC value between 600 and 1000 μm2/s;

  • For soft-tissue disease, RECIST version 1.1 criteria meeting requirements for PD (≥ 20% increase in the sum of L-Di of TLs, taking as reference the smallest sum L-Di recorded since the treatment started, or the appearance of ≥1 new lesion(s))

 
Metabolic responseDescriptionRACsDescription
Complete metabolic response Uptake less than or equal to liver activity in BM sites and FLs previously involved, including PSD and EMD (DS 1-3) RAC1 Highly likely to be responding:
  • Return of normal fat containing marrow in areas previously infiltrated by FLs or DD;

  • Unequivocal decrease in number or size of FLs;

  • Conversion of a packed BM infiltrate into discrete nodules, with unequivocal decrease in tumor load in the respective BM space;

  • Decreasing soft tissue associated with bone disease;

  • Emergence of intra- or peritumoral fat within/around FLs (fat dot or halo sign);

  • Previously evident lesion shows increase in ADC from ≤1400 μm2/s to >1400 μm2/s;

  • ≥40% increase in ADC from baseline, with corresponding decrease in normalized high b-value signal intensity, and morphologic findings consistent with stable or responding disease;

  • For soft-tissue disease, RECIST version 1.1 criteria for CR (disappearance of all TLs)/PR (≥30% decrease in the sum of L-Di of TLs, taking as reference the baseline sum L-Di)

 
Partial metabolic response Decrease in number and/or activity of BM/FLs present at baseline, but persistence of lesion(s) with uptake more than liver activity (DS 4-5) RAC2 Likely to be responding:
  • Evidence of improvement but not enough to fulfill criteria for RAC1, for example, slight decrease in number/size of FLs, previously evident lesions showing increases in ADC from ≤1000 μm2/s to <1400 μm2/s, a 25% to 40% increase in ADC from baseline with corresponding decrease in b-value signal intensity, and morphologic findings consistent with stable or responding disease;

  • For soft-tissue disease, RECIST version 1.1 criteria not meeting requirements for PR

 
Stable metabolic disease No significant change in BM/FLs compared with baseline RAC3 No change:
  • No observable change

 
Progressive metabolic disease New FLs consistent with MM compared with baseline RAC4 Likely to be progressing:
  • Evidence of worsening disease, but not enough to fulfill criteria for RAC5;

  • Equivocal appearance of new lesion(s);

  • No change in size but increasing signal intensity on high b-value images (with ADC values <1400 μm2/s) consistent with possible disease progression;

  • Relapsed disease: reemergence of lesion(s) that previously disappeared or enlargement of lesion(s) that had partially regressed/stabilized with prior treatments;

  • Soft tissue in spinal canal causing narrowing not associated with neurologic findings and not requiring radiation therapy;

  • For soft-tissue disease, RECIST version 1.1 criteria not meeting requirements for PD

 
  RAC5 Highly likely to be progressing:
  • New critical fracture(s)/cord compression requiring radiation therapy/surgical intervention; only if confirmed as malignant with MRI signal characteristics;

  • Unequivocal new focal (5-10 mm)/diffuse area(s) of infiltration in regions of previously normal marrow;

  • Unequivocal increase in number/size of FLs;

  • Evolution of FLs to diffuse neoplastic pattern;

  • Appearance/increasing soft tissue associated with bone disease;

  • New lesions/regions of high signal intensity on high b-value images with ADC value between 600 and 1000 μm2/s;

  • For soft-tissue disease, RECIST version 1.1 criteria meeting requirements for PD (≥ 20% increase in the sum of L-Di of TLs, taking as reference the smallest sum L-Di recorded since the treatment started, or the appearance of ≥1 new lesion(s))

 

Adapted from Zamagni et al80 and Messiou et al.82 

CR, complete response; L-Di, longest diameter; PD, progressive disease; PR, partial response; TL, target lesion.

Furthermore, several studies (both in NDMM and RRMM, including early experiences with novel immunotherapies) have demonstrated variability between imaging and BM MRD, thus proposing their complementary integration to improve prognostic stratification. Overall, negativity of both 18F-FDG–PET/CT and BM MRD was found to be predictive of superior outcomes.77,81,83-89 

DW imaging (DWI) represents a modern MRI protocol measuring the movement of water molecules within tissues, resulting inversely correlated to its cellular density. Myeloma response assessment and diagnosis system (MY-RADS) guidelines have proposed a standardization of acquisition, interpretation, and reporting of DW-MRI in MM, including its use in response assessment. This protocol uses field strength of 1.5 to 3 Tesla with a WB-FOV (skull vertex to knees) and section thickness of ∼4 to 5 mm. Scanning time ranges from 40 to 60 minutes. It relies on both conventional morphological sequences and functional ones, without need for contrast agents. The formers include T1-weighted, T2-weighted, and short tau inversion recovery sequences, whereas the latter are represented by DWI and T1-weighted sequences with Dixon technique for fat suppression. DWI is based on qualitative parameters (particularly the comparison with adjacent muscle using high b-value images, generally 800-900 s/mm2) and semiquantitative approaches, particularly the apparent diffusion coefficient (ADC).82 This parameter has been significantly related to the grade of histological infiltration by malignant PCs and microvascular density.90 

Normal BM is generally characterized by ADC values <600 to 700 μm2/s, due the presence of fat yellow marrow (with decreasing values in older people). Conversely, values of viable tumor are generally between 700 and 1400 μm2/s (Figure 2). Four disease patterns have been described: diffuse, micronodular, focal, and focal on diffuse (Figure 1; Table 3).90 As well as evaluation of ADC maps regarding BM infiltration and FLs, a volumetric parameter named total diffusion volume (TDV), calculated by measuring the voxels with abnormal ADC values, has been proposed to estimate tumor burden.91 

Figure 2.

Relation between ADC values, signal intensity in DWI sequences, and BM cellular density during disease course and response to therapy. MGUS, monoclonal gammopathy of undetermined significance. Adapted from Dutoit et al92 and Ormond et al.93 

Figure 2.

Relation between ADC values, signal intensity in DWI sequences, and BM cellular density during disease course and response to therapy. MGUS, monoclonal gammopathy of undetermined significance. Adapted from Dutoit et al92 and Ormond et al.93 

Close modal
Table 3.

DW-MRI disease patterns

Disease patternDescription
Diffuse Diffuse decreased signal intensity in T1-weighted sequences and increased signal intensity in T2-weighted, STIR, and high b-value DW sequences, with ADC values >600 to 700 μm2/s 
Micronodular Widespread nodular areas <5 mm with preserved normal marrow in between 
Focal Lesions ≥5 mm with decreased signal in T1-weighted sequences, increased signal in T2-weighted and STIR sequences, hyperintense to background muscle in high b-value images, compatible ADC values (600-1400 μm2/s) 
Focal on diffuse Coexistence of focal and DD 
Disease patternDescription
Diffuse Diffuse decreased signal intensity in T1-weighted sequences and increased signal intensity in T2-weighted, STIR, and high b-value DW sequences, with ADC values >600 to 700 μm2/s 
Micronodular Widespread nodular areas <5 mm with preserved normal marrow in between 
Focal Lesions ≥5 mm with decreased signal in T1-weighted sequences, increased signal in T2-weighted and STIR sequences, hyperintense to background muscle in high b-value images, compatible ADC values (600-1400 μm2/s) 
Focal on diffuse Coexistence of focal and DD 

Adapted from Messiou et al.82 

STIR, short tau inversion recovery.

In patients responding to therapies, MRI can depict several signal changes. Indeed, decrease in number and size of FLs, reduced contrast uptake, modifications of BM pattern, increase of signal intensity in T1-weighted sequences due to fat conversion around or within the lesions (“fat dot” and “halo sign”) and decrease of signal intensity in T2-weighted or short tau inversion recovery can be shown by conventional MRI.94 However, WB-DW-MRI displays additional changes, as it integrates Dixon and DWI sequences. The former may highlight fat reconversion, as responding patients are characterized by higher increase of BM fat fraction (FF) than nonresponders.95,96 Furthermore, treatment response is characterized by significant changes of ADC values: tumor necrosis, microbleeding, and edema lead to reduction of cellular density and subsequent increased ADC values in early evaluations, whereas fat reconversion causes decrease of values in later assessments (Figure 2). Heterogeneous cutoff values have been proposed, with a great limitation represented by use of diverse b-values. MY-RADS guidelines have defined 5 response assessment categories (RACs), aimed at evaluating the probability of response or progression,82 a different approach from the categorical responses of IMPeTUs (Table 2). Usefulness and applicability of MY-RADS have been confirmed both in clinical trials and in the real-world setting.97,98 

WB-DW-MRI at staging

Use of DW-MRI at baseline staging has been correlated to pooled sensitivity and specificity of 86% and 63%, respectively, with significant superiority over conventional MRI.99 WB-DW-MRI has been compared with WBLDCT, showing a good concordance regarding evaluation of focal involvement, number, and distribution of FLs.100 

Furthermore, comparisons to 18F-FDG–PET/CT showed higher sensitivity of WB-DW-MRI regarding detection of FLs (with significant superiority in all anatomic regions, except the skull) and PSD, similar sensitivity in detecting EMD and discordant data regarding assessment of DD.101-103 Inter alia, “iTIMM” (to our knowledge, the only prospective trial published to date in this setting) has demonstrated superiority of WB-DW-MRI compared with 18F-FDG–PET/CT for detection of both FLs (83% vs 60%) and DD (82% vs 17%). Specifically, WB-DW-MRI was significantly more sensitive than the nuclear technique in all skeletal districts, apart from ribs, scapulae, and clavicles.104 

Early results from our prospective experience have confirmed the superiority of WB-DW-MRI over 18F-FDG–PET/CT in detecting FLs (76% vs 54%) and PSD (30% vs 20%), whereas a slight concordance in detecting DD was observed.105 However, a retrospective study observed that treatment decisions were not statistically different if combining the 2 techniques, concluding that both are appropriate for initial staging.104 

WB-DW-MRI in prognostic stratification

Studies regarding the prognostic role of baseline conventional MRI have demonstrated a significant impact of high number of FLs (>7 with AS-MRI, >25 with WB-MRI) and DD.11,106 

Conversely, studies with DW-MRI have highlighted that the presence of ≥3 FLs with product of perpendicular diameters >5 cm2 is associated to inferior PFS and OS, independently from genetic risk, R-ISS, and presence of EMD. The number of FLs lost its prognostic impact after adjustment for size of FLs, emphasizing the role of high tumor burden.107 Furthermore, the presence of DD has been related to several markers of prognosis and disease burden, including high R-ISS, high-risk cytogenetics, greater BM-PC infiltration, higher M-protein concentration, and lower hemoglobin level.104,105 A significant prognostic role has also been reported for elevated TDV and ADC values.108-110 

WB-DW-MRI in response assessment

Early studies with conventional MRI had shown a prognostic significance of FLs resolution after therapy,11,111 but high rates of false-positive results (due to the persistence of visible necrotic lesions on morphological sequences) have been reported, and recent trials failed to show a significant correlation with patients’ outcome.32 

Conversely, several studies have shown a significant association between the increase of ADC values after treatment and the presence of a biochemical response.112,113 Furthermore, early modifications of FF metrics (after 1 cycle of induction therapy) and a progressive decrease of TDV, resulting directly proportional to the depth of hematologic response, have been observed.114,115 

Use of DW-MRI in response assessment has been related to sensitivity and specificity of 78% and 73%, respectively, by a recent meta-analysis.99 Another meta-analysis has demonstrated a non-significant improvement of sensitivity (93% vs 74%), with similar specificity, as compared with conventional WB-MRI without DWI.116 Response assessment using MY-RADS guidelines has been proven prognostic: achievement of RAC1 after ASCT and sustained RAC1 after 1 year have been related to significant improvement of PFS and OS.98,117 Furthermore, significant correlation between greater decrease of relative FF and improved PFS was shown, thereby proposing a complementarity with conventional RACs.118 

Comparison to 18F-FDG–PET/CT in response assessment highlighted good concordance.105 However, WB-DW-MRI identifies more residual FLs after treatment, though some FLs are only PET positive, thereby proposing a complementarity of the 2 techniques.84,89 A recent meta-analysis has compared 18F-FDG–PET/CT and WB-MRI (with DWI included in 5/12 studies), demonstrating significantly superior specificity of 18F-FDG–PET/CT (81% vs 56%) and nonsignificantly superior sensitivity of WB-MRI (90% vs 66%).116 Nevertheless, the persistence of positive 18F-FDG–PET/CT after induction therapy or after ASCT was correlated to a more significant impact on PFS than absence of response by WB-DW-MRI.119 

Recent studies have also proposed an integration of response assessment by DW-MRI and BM MRD evaluation, showing better outcomes for double-negative patients than patients with positive MRD/imaging and double-positives.84,89,98 Improved outcome in patients with NDMM/RRMM receiving consolidation strategies because of imaging (DW-MRI or 18F-FDG–PET/CT) or MRD positivity was also shown.120Table 4 compares 18F-FDG–PET/CT and WB-DW-MRI.

Table 4.

Main features, advantages, and disadvantages of18F-FDG–PET/CT and WB-DW-MRI in MM

18F-FDG–PET/CTWB-DW-MRI
Scanning time 15-20 minutes (60 minutes after radiotracer infusion) 40-60 minutes 
Radiation exposure 10-25 mSv None 
Diffuse BM involvement Lower sensitivity Higher sensitivity 
Detection of FLs Lower sensitivity Higher sensitivity 
Detection of PSD Lower sensitivity Higher sensitivity 
Detection of EMD Gold-standard technique Less explored role (apparent similar sensitivity) 
Prognostic features >3 FLs, high SUVmax, other quantitative parameters (MTV, TLG, IBI, and PBI) DD, >3 large (>5 cm2) FLs, high TDV, high ADC values 
Role in response assessment Gold-standard technique (IMWG); recent IMPeTUs for definition of metabolic responses (prognostic role of DS <4) Recommended technique for British guidelines (MY-RADS criteria: prognostic role of RAC1) 
18F-FDG–PET/CTWB-DW-MRI
Scanning time 15-20 minutes (60 minutes after radiotracer infusion) 40-60 minutes 
Radiation exposure 10-25 mSv None 
Diffuse BM involvement Lower sensitivity Higher sensitivity 
Detection of FLs Lower sensitivity Higher sensitivity 
Detection of PSD Lower sensitivity Higher sensitivity 
Detection of EMD Gold-standard technique Less explored role (apparent similar sensitivity) 
Prognostic features >3 FLs, high SUVmax, other quantitative parameters (MTV, TLG, IBI, and PBI) DD, >3 large (>5 cm2) FLs, high TDV, high ADC values 
Role in response assessment Gold-standard technique (IMWG); recent IMPeTUs for definition of metabolic responses (prognostic role of DS <4) Recommended technique for British guidelines (MY-RADS criteria: prognostic role of RAC1) 

IBI, intensity of bone involvement; PBI, percentage of bone involvement; TLG, total lesion glycolysis.

Dynamic contrast-enhanced (DCE) MRI is a functional protocol that allows to study tumor angiogenesis and microcirculation. It is based on high-temporal resolution repeated scanning before, during, and after gadolinium administration, using T1-weighted sequences of dorsolumbar spine. Time-intensity curves represent changes in local perfusion over time, and are based on the assessment of 2 main parameters: amplitude A, reflecting blood volume, and rate constant Kep, reflecting vessel wall permeability. Symptomatic MM is usually characterized by a steep wash-in and first pass (due to immediate flow from intravascular to interstitial space because of marked microvascular density and vessel permeability) and rapid washout (as high cellular density and small interstitial space cause a retrograde flow toward intravascular space). Patients rarely show steep but continuous wash-in, reflecting the persistence of large interstitial space.92,121 

DCE-MRI displays a progressive increment of microcirculation from monoclonal gammopathy of undetermined significance to SMM to MM, with gradual increase of peak enhancement intensity, and decrease of time to peak enhancement intensity.122 

Furthermore, higher amplitude A was proven a risk factor for progression of SMM to symptomatic MM, and both amplitude A and Kep were found to have a significant prognostic impact in patients with MM.123-126 

Patients responding to therapy usually show significant decrease of wash-in slope and absolute enhancement, likely due to destruction of tumor vascularization. However, a steep wash-in and subsequent plateau may be observed in responders, reflecting increased vascularization due to hematopoiesis regeneration, following anti-MM treatment and stimulating agents.127,128 Moreover, lower values of maximal percentage of BM enhancement, amplitude A, and Kep have been observed in patients responding to therapy.124,129 A recent study has proposed a “combined skeletal score,” based on the integration of morphological and functional MRI data, from both DCE and DWI protocols, to enhance response assessment, particularly for complete response confirmation and prediction of relapse in apparently good-responding patients.128 

Though encouraging, DCE-MRI use is limited by lack of clinical validation and standardization. Furthermore, it requires adjustment of results for variables impacting BM vascularization, including body mass index, age, and sex. Another limit is represented by gadolinium administration in patients with severe renal impairment, due to increased risk of nephrogenic systemic sclerosis.130 

18F-FDG–PET/MRI represents a hybrid imaging technique currently being evaluated in PC dyscrasias.

A retrospective comparison between 18F-FDG–PET, WB-MRI and 18F-FDG–PET/MRI including DWI or DCE protocols showed superiority of 18F-FDG–PET/MRI over PET alone, regarding detection of both FLs (sensitivity, specificity and accuracy of 93%, 97%, and 95%) and DD (sensitivity, specificity and accuracy of 98%, 66%, and 79%), without improvement by adding DCE.131 A prospective trial enrolling patients with NDMM compared the diagnostic performance of this hybrid technique to PET and MRI separately, demonstrating similar accuracy for detection of FLs and DD in symptomatic patients, but superiority of MRI in SMM (allowing detection of FLs in 22% of patients).132 

Furthermore, 18F-FDG–PET/MRI was recently evaluated in response assessment: concomitant negativity of PET (DS ≤3) and WB-DW-MRI (RAC1) after ASCT was related to significantly superior PFS than persistent positivity in 1 technique, particularly in case of MRI positivity.133 

The main advantages of 18F-FDG–PET/MRI include the concomitant evaluation of morphology, BM cellular density, vascularization, and metabolic activity. However, it is scarcely available, expensive, and requires double expertise.

18F-FDG–PET/CT is currently considered the standard imaging technique in MM, as it combines morphologic and functional data. Indeed, its CT portion represents a recommended technique for MBD diagnosis, whereas the metabolic counterpart has a well-recognized role in prognostic stratification and response assessment, representing the recommended technique in this setting. However, this imaging technique harbors some limitations, with various causes of false-positive or -negative results. Recent years have been characterized by the introduction of novel functional techniques, first among all WB-DW-MRI. Its use in MM appears very promising, as various studies comparing it to 18F-FDG–PET/CT at staging have demonstrated higher sensitivity regarding detection of FLs, PSD, and DD, and similar sensitivity regarding EMD. Furthermore, a significant prognostic role in response assessment was shown, thereby overcoming the major limitation of conventional MRI. Indeed, British guidelines recommend its use as gold-standard imaging technique, and propose it as an alternative to 18F-FDG–PET/CT in response evaluation,134 though IMWG guidelines do not recommend it in response assessment. However, drafting of novel guidelines regarding response assessment is ongoing and it will likely advocate use of only functional imaging techniques (without further need for assessment of plasmacytomas size through conventional morphological techniques), and propose DW-MRI as an alternative to 18F-FDG–PET/CT. Notably, it should be highlighted that use of WB-DW-MRI is currently limited by slight availability and complex evaluation requiring specific expertise.

Furthermore, new software and algorithms, even using radiomics and artificial intelligence (in its declinations of machine and deep learning) are in development with promising results. The main aims are simplification, enhancement of reproducibility, improvement of diagnostic performance, noninvasive prediction of patients’ outcome and assistance to clinical choices.135-139 

Future studies will have to further elucidate the complementarity of different functional imaging techniques, to investigate whether one of them might become a new gold standard or they might be alternatively used, and to assess and standardize the role of artificial intelligence tools in MM.

The authors acknowledge the Departments of Radiology and Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, for providing the images used in Figure 1.

Contribution: E.Z. designed research, discussed data, supervised the project, critically reviewed the manuscript, and approved the final version of the manuscript for publication; and M.T. performed research, discussed data, and drafted the original version of the manuscript.

Conflict-of-interest disclosure: E.Z. reports honoraria from Janssen, Bristol Myers Squibb, Amgen, and Takeda. M.T. declares no competing financial interests.

Correspondence: Elena Zamagni, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia “Seràgnoli,” Via Massarenti 9, 40138 Bologna, Italy; email: e.zamagni@unibo.it.

1.
Landgren
O
,
Kyle
RA
,
Pfeiffer
RM
, et al
.
Monoclonal gammopathy of undetermined significance (MGUS) consistently precedes multiple myeloma: a prospective study
.
Blood
.
2009
;
113
(
22
):
5412
-
5417
.
2.
Kyle
RA
,
Gertz
MA
,
Witzig
TE
, et al
.
Review of 1027 patients with newly diagnosed multiple myeloma
.
Mayo Clin Proc
.
2003
;
78
(
1
):
21
-
33
.
3.
Terpos
E
,
Berenson
J
,
Raje
N
,
Roodman
GD
.
Management of bone disease in multiple myeloma
.
Expert Rev Hematol
.
2014
;
7
(
1
):
113
-
125
.
4.
Rajkumar
SV
,
Dimopoulos
MA
,
Palumbo
A
, et al
.
International Myeloma Working Group updated criteria for the diagnosis of multiple myeloma
.
Lancet Oncol
.
2014
;
15
(
12
):
e538
-
e548
.
5.
Hillengass
J
,
Fechtner
K
,
Weber
MA
, et al
.
Prognostic significance of focal lesions in whole-body magnetic resonance imaging in patients with asymptomatic multiple myeloma
.
J Clin Oncol
.
2010
;
28
(
9
):
1606
-
1610
.
6.
Kastritis
E
,
Moulopoulos
LA
,
Terpos
E
,
Koutoulidis
V
,
Dimopoulos
MA
.
The prognostic importance of the presence of more than one focal lesion in spine MRI of patients with asymptomatic (smoldering) multiple myeloma
.
Leukemia
.
2014
;
28
(
12
):
2402
-
2403
.
7.
Rosiñol
L
,
Beksac
M
,
Zamagni
E
, et al
.
Expert review on soft-tissue plasmacytomas in multiple myeloma: definition, disease assessment and treatment considerations
.
Br J Haematol
.
2021
;
194
(
3
):
496
-
507
.
8.
Bladé
J
,
Beksac
M
,
Caers
J
, et al
.
Extramedullary disease in multiple myeloma: a systematic literature review
.
Blood Cancer J
.
2022
;
12
(
3
):
45
.
9.
Hillengass
J
,
Moulopoulos
LA
,
Delorme
S
, et al
.
Whole-body computed tomography versus conventional skeletal survey in patients with multiple myeloma: a study of the International Myeloma Working Group
.
Blood Cancer J
.
2017
;
7
(
8
):
e599
.
10.
Hillengass
J
,
Usmani
S
,
Rajkumar
SV
, et al
.
International Myeloma Working Group consensus recommendations on imaging in monoclonal plasma cell disorders [published correction appears in Lancet Oncol. 2019;20(7):e346]
.
Lancet Oncol
.
2019
;
20
(
6
):
e302
-
e312
.
11.
Walker
R
,
Barlogie
B
,
Haessler
J
, et al
.
Magnetic resonance imaging in multiple myeloma: diagnostic and clinical implications
.
J Clin Oncol
.
2007
;
25
(
9
):
1121
-
1128
.
12.
Zamagni
E
,
Nanni
C
,
Patriarca
F
, 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
(
1
):
50
-
55
.
13.
Spinnato
P
,
Bazzocchi
A
,
Brioli
A
, et al
.
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
(
12
):
4013
-
4018
.
14.
Regelink
JC
,
Minnema
MC
,
Terpos
E
, et al
.
Comparison of modern and conventional imaging techniques in establishing multiple myeloma-related bone disease: a systematic review
.
Br J Haematol
.
2013
;
162
(
1
):
50
-
61
.
15.
Kumar
S
,
Paiva
B
,
Anderson
KC
, et al
.
International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma
.
Lancet Oncol
.
2016
;
17
(
8
):
e328
-
e346
.
16.
Cavo
M
,
Terpos
E
,
Nanni
C
, et al
.
Role of 18F-FDG PET/CT in the diagnosis and management of multiple myeloma and other plasma cell disorders: a consensus statement by the International Myeloma Working Group
.
Lancet Oncol
.
2017
;
18
(
4
):
e206
-
e217
.
17.
Nanni
C
,
Deroose
CM
,
Balogova
S
, et al
.
EANM guidelines on the use of [18F]FDG PET/CT in diagnosis, staging, prognostication, therapy assessment, and restaging of plasma cell disorders
.
Eur J Nucl Med Mol Imaging
.
2024
;
52
(
1
):
171
-
192
.
18.
Adams
MC
,
Turkington
TG
,
Wilson
JM
,
Wong
TZ
.
A systematic review of the factors affecting accuracy of SUV measurements [published correction appears in Am J Roentgenol 2010;195(4):1043]
.
Am J Roentgenol
.
2010
;
195
(
2
):
310
-
320
.
19.
Takahashi
MES
,
Lorand-Metze
I
,
de Souza
CA
, et al
.
Metabolic volume measurements in multiple myeloma
.
Metabolites
.
2021
;
11
(
12
):
875
.
20.
Fonti
R
,
Larobina
M
,
Del Vecchio
S
, et al
.
Metabolic tumor volume assessed by 18F-FDG PET/CT for the prediction of outcome in patients with multiple myeloma
.
J Nucl Med
.
2012
;
53
(
12
):
1829
-
1835
.
21.
McDonald
JE
,
Kessler
MM
,
Gardner
MW
, et al
.
Assessment of total lesion glycolysis by 18F FDG PET/CT significantly improves prognostic value of GEP and ISS in myeloma
.
Clin Cancer Res
.
2017
;
23
(
8
):
1981
-
1987
.
22.
Terao
T
,
Machida
Y
,
Tsushima
T
, et al
.
Pre-treatment metabolic tumour volume and total lesion glycolysis are superior to conventional positron-emission tomography/computed tomography variables for outcome prediction in patients with newly diagnosed multiple myeloma in clinical practice
.
Br J Haematol
.
2020
;
191
(
2
):
223
-
230
.
23.
Terao
T
,
Machida
Y
,
Hirata
K
, et al
.
Prognostic impact of metabolic heterogeneity in patients with newly diagnosed multiple myeloma using 18F-FDG PET/CT
.
Clin Nucl Med
.
2021
;
46
(
10
):
790
-
796
.
24.
Rasche
L
,
Angtuaco
E
,
McDonald
JE
, et al
.
Low expression of hexokinase-2 is associated with false-negative FDG-positron emission tomography in multiple myeloma
.
Blood
.
2017
;
130
(
1
):
30
-
34
.
25.
von Hinten
J
,
Kircher
M
,
Dierks
A
, et al
.
Molecular imaging in multiple myeloma-novel PET radiotracers improve patient management and guide therapy
.
Front Nucl Med
.
2022
;
2
:
801792
.
26.
Talarico
M
,
Barbato
S
,
Cattabriga
A
, et al
.
Diagnostic innovations: advances in imaging techniques for diagnosis and follow-up of multiple myeloma
.
J Bone Oncol
.
2025
;
51
:
100669
.
27.
Nanni
C
,
Versari
A
,
Chauvie
S
, et al
.
Interpretation criteria for FDG PET/CT in multiple myeloma (IMPeTUs): final results. IMPeTUs (Italian myeloma criteria for PET USe)
.
Eur J Nucl Med Mol Imaging
.
2018
;
45
(
5
):
712
-
719
.
28.
Siontis
B
,
Kumar
S
,
Dispenzieri
A
, et al
.
Positron emission tomography-computed tomography in the diagnostic evaluation of smoldering multiple myeloma: identification of patients needing therapy
.
Blood Cancer J
.
2015
;
5
(
10
):
e364
.
29.
Zamagni
E
,
Patriarca
F
,
Nanni
C
, 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
(
23
):
5989
-
5995
.
30.
Bartel
TB
,
Haessler
J
,
Brown
TL
, et al
.
F18-fluorodeoxyglucose positron emission tomography in the context of other imaging techniques and prognostic factors in multiple myeloma
.
Blood
.
2009
;
114
(
10
):
2068
-
2076
.
31.
van Lammeren-Venema
D
,
Regelink
JC
,
Riphagen
II
,
Zweegman
S
,
Hoekstra
OS
,
Zijlstra
JM
.
18F-fluoro-deoxyglucose positron emission tomography in assessment of myeloma-related bone disease: a systematic review
.
Cancer
.
2012
;
118
(
8
):
1971
-
1981
.
32.
Moreau
P
,
Attal
M
,
Caillot
D
, 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
(
25
):
2911
-
2918
.
33.
Sager
S
,
Ergül
N
,
Ciftci
H
,
Cetin
G
,
Güner
SI
,
Cermik
TF
.
The value of FDG PET/CT in the initial staging and bone marrow involvement of patients with multiple myeloma
.
Skeletal Radiol
.
2011
;
40
(
7
):
843
-
847
.
34.
Maggialetti
N
,
Ferrari
C
,
Nappi
AG
, et al
.
Is whole body low dose CT still necessary in the era of 18F-FDG PET/CT for the assessment of bone disease in multiple myeloma patients?
.
J Nucl Med
.
2020
;
23
(
3
):
264
-
271
.
35.
Shortt
CP
,
Gleeson
TG
,
Breen
KA
, et al
.
Whole-body MRI versus PET in assessment of multiple myeloma disease activity
.
Am J Roentgenol
.
2009
;
192
(
4
):
980
-
986
.
36.
Lecouvet
FE
,
Boyadzhiev
D
,
Collette
L
, et al
.
MRI versus 18F-FDG-PET/CT for detecting bone marrow involvement in multiple myeloma: diagnostic performance and clinical relevance
.
Eur Radiol
.
2020
;
30
(
4
):
1927
-
1937
.
37.
Lu
YY
,
Chen
JH
,
Lin
WY
, et al
.
FDG PET or PET/CT for detecting intramedullary and extramedullary lesions in multiple myeloma: a systematic review and meta-analysis
.
Clin Nucl Med
.
2012
;
37
(
9
):
833
-
837
.
38.
Zamagni
E
,
Nanni
C
,
Gay
F
, et al
.
18F-FDG PET/CT focal, but not osteolytic, lesions predict the progression of smoldering myeloma to active disease
.
Leukemia
.
2016
;
30
(
2
):
417
-
422
.
39.
Han
S
,
Woo
S
,
Kim
YI
,
Yoon
DH
,
Ryu
JS
.
Prognostic value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography in newly diagnosed multiple myeloma: a systematic review and meta-analysis
.
Eur Radiol
.
2021
;
31
(
1
):
152
-
162
.
40.
Li
Q
,
Hu
L
,
Charwudzi
A
,
Zhu
W
,
Meng
Y
,
Zhai
Z
.
Prognostic value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography at diagnosis in untreated multiple myeloma patients: a systematic review and meta-analysis
.
Clin Exp Med
.
2023
;
23
(
1
):
31
-
43
.
41.
Usmani
SZ
,
Mitchell
A
,
Waheed
S
, et al
.
Prognostic implications of serial 18-fluoro-deoxyglucose emission tomography in multiple myeloma treated with total therapy 3
.
Blood
.
2013
;
121
(
10
):
1819
-
1823
.
42.
Zamagni
E
,
Nanni
C
,
Mancuso
K
, et al
.
PET/CT improves the definition of complete response and allows to detect otherwise unidentifiable skeletal progression in multiple myeloma
.
Clin Cancer Res
.
2015
;
21
(
19
):
4384
-
4390
.
43.
Michaud-Robert
AV
,
Zamagni
E
,
Carlier
T
, et al
.
Glucose metabolism quantified by SUVmax on baseline FDG-PET/CT predicts survival in newly diagnosed multiple myeloma patients: combined harmonized analysis of two prospective phase III trials
.
Cancers (Basel)
.
2020
;
12
(
9
):
2532
.
44.
Jung
SH
,
Kwon
SY
,
Min
JJ
, et al
.
18F-FDG PET/CT is useful for determining survival outcomes of patients with multiple myeloma classified as stage II and III with the Revised International Staging System
.
Eur J Nucl Med Mol Imaging
.
2019
;
46
(
1
):
107
-
115
.
45.
Cho
HJ
,
Jung
SH
,
Jo
JC
, et al;
Korean Multiple Myeloma Working Party
.
Development of a new risk stratification system for patients with newly diagnosed multiple myeloma using R-ISS and 18F-FDG PET/CT
.
Blood Cancer J
.
2021
;
11
(
12
):
190
.
46.
Abe
Y
,
Narita
K
,
Kobayashi
H
, et al
.
Pretreatment 18F-FDG PET/CT combined with quantification of clonal circulating plasma cells as a potential risk model in patients with newly diagnosed multiple myeloma
.
Eur J Nucl Med Mol Imaging
.
2019
;
46
(
6
):
1325
-
1333
.
47.
Montefusco
V
,
Gay
F
,
Spada
S
, et al
.
Outcome of paraosseous extra-medullary disease in newly diagnosed multiple myeloma patients treated with new drugs
.
Haematologica
.
2020
;
105
(
1
):
193
-
200
.
48.
He
J
,
Yue
X
,
He
D
, et al
.
Multiple extramedullary-bone related and/or extramedullary extraosseous are independent poor prognostic factors in patients with newly diagnosed multiple myeloma
.
Front Oncol
.
2021
;
11
:
668099
.
49.
Jiménez-Segura
R
,
Rosiñol
L
,
Cibeira
MT
, et al
.
Paraskeletal and extramedullary plasmacytomas in multiple myeloma at diagnosis and at first relapse: 50-years of experience from an academic institution
.
Blood Cancer J
.
2022
;
12
(
9
):
135
.
50.
Alberge
JB
,
Kraeber-Bodéré
F
,
Jamet
B
, et al
.
Molecular signature of 18F-FDG PET biomarkers in newly diagnosed multiple myeloma patients: a genome-wide transcriptome analysis from the CASSIOPET study
.
J Nucl Med
.
2022
;
63
(
7
):
1008
-
1013
.
51.
Takahashi
MES
,
Mosci
C
,
Souza
EM
, et al
.
Proposal for a quantitative 18F-FDG PET/CT metabolic parameter to assess the intensity of bone involvement in multiple myeloma
.
Sci Rep
.
2019
;
9
(
1
):
16429
.
52.
Ikeda
D
,
Terao
T
,
Oura
M
, et al
.
Analysis of baseline circulating tumor cells integrated with PET/CT findings in transplant-ineligible multiple myeloma
.
Blood Adv
.
2024
;
8
(
1
):
37
-
46
.
53.
Sachpekidis
C
,
Enqvist
O
,
Ulén
J
, et al
.
Application of an artificial intelligence-based tool in [18F]FDG PET/CT for the assessment of bone marrow involvement in multiple myeloma
.
Eur J Nucl Med Mol Imaging
.
2023
;
50
(
12
):
3697
-
3708
.
54.
Lapa
C
,
Lückerath
K
,
Malzahn
U
, et al
.
18 FDG-PET/CT for prognostic stratification of patients with multiple myeloma relapse after stem cell transplantation
.
Oncotarget
.
2014
;
5
(
17
):
7381
-
7391
.
55.
Jamet
B
,
Bailly
C
,
Carlier
T
, et al
.
Added prognostic value of FDG-PET/CT in relapsing multiple myeloma patients
.
Leuk Lymphoma
.
2019
;
60
(
1
):
222
-
225
.
56.
Fouquet
G
,
Wartski
M
,
Dechmi
A
, et al
.
Prognostic value of FDG-PET/CT parameters in patients with relapse/refractory multiple myeloma before anti-CD38 based therapy
.
Cancers (Basel)
.
2021
;
13
(
17
):
4323
.
57.
Zukovs
R
,
Antke
C
,
Mamlins
E
, et al
.
18F-FDG-PET/CT in relapsed multiple myeloma: are prognostic thresholds different from first-line therapy?
.
BMC Med Imaging
.
2022
;
22
(
1
):
63
.
58.
Moreau
P
,
Mateos
MV
,
Goldschmidt
H
, et al
.
Outcomes of patients with extramedullary disease in triple-class exposed relapsed/refractory multiple myeloma from the LocoMMotion + MoMMent studies
.
Blood
.
2023
;
142
(
suppl 1
):
4698
.
59.
Tamariz-Amador
LE
,
Alfonso-Pierola
A
,
Huerga-Dominguez
S
, et al
.
OA-11 utility of 18FDG-PET/CT for risk prediction in relapsed/refractory multiple myeloma patients undergoing CAR-T cell therapy: analysis of baseline and early assessment scans after one and three months
.
Clin Lymphoma Myeloma Leuk
.
2023
;
23
(
suppl 2
):
S7
.
60.
Arnulf
B
,
Vercellino
L
,
Caillot
D
, et al
.
Prognostic value of positron emission tomography/computed tomography in multiple myeloma patients treated with Ide-Cel CAR-T cell therapy: preliminary results of a real-world IFM study from the Descar-T registry (CAR MY PET)
.
Blood
.
2024
;
144
(
suppl 1
):
4765
.
61.
Zanwar
S
,
Sidana
S
,
Shune
L
, et al
.
Impact of extramedullary multiple myeloma on outcomes with idecabtagene vicleucel
.
J Hematol Oncol
.
2024
;
17
(
1
):
42
.
62.
Freeman
CL
,
Noble
J
,
Menges
M
, et al
.
Tumor burden quantified by soluble B-cell maturation antigen and metabolic tumor volume determine myeloma CAR-T outcomes
.
Blood
.
2025
;
145
(
15
):
1645
-
1657
.
63.
Zugasti
I
,
Tormo-Ratera
M
,
Oliver-Caldés
A
, et al
.
Clinical impact of [18F]FDG-PET/CT in ARI0002h treatment, a CAR-T against BCMA for relapsed/refractory multiple myeloma
.
Blood Adv
.
2025
;
9
(
3
):
571
-
582
.
64.
Amoozgar
B
,
Bangolo
A
,
Phull
P
, et al
.
Impact of extramedullary disease on survival outcomes in multiple myeloma patients treated with ciltacabtagene autoleucel and idecabtagene vicleucel: a comparative analysis
.
Transplant Cell Ther
.
2025
;
31
(
2
):
S237
-
S238
.
65.
Park
S
,
Lee
SJ
,
Chang
WJ
, et al
.
Positive correlation between baseline PET or PET/CT findings and clinical parameters in multiple myeloma patients
.
Acta Haematol
.
2014
;
131
(
4
):
193
-
199
.
66.
Zhou
X
,
Dierks
A
,
Kertels
O
, et al
.
The link between cytogenetics/genomics and imaging patterns of relapse and progression in patients with relapsed/refractory multiple myeloma: a pilot study utilizing 18F-FDG PET/CT
.
Cancers (Basel)
.
2020
;
12
(
9
):
2399
.
67.
Sachpekidis
C
,
Merz
M
,
Raab
MS
, et al
.
The prognostic significance of [18F]FDG PET/CT in multiple myeloma according to novel interpretation criteria (IMPeTUs)
.
EJNMMI Res
.
2021
;
11
(
1
):
100
.
68.
Deng
S
,
Zhang
B
,
Zhou
Y
, et al
.
The role of 18F-FDG PET/CT in multiple myeloma staging according to IMPeTUs: comparison of the Durie-Salmon Plus and other staging systems
.
Contrast Media Mol Imaging
.
2018
;
2018
:
4198673
.
69.
Marchiori
S
,
Cousin
F
,
Papadopoulos
I
, et al
.
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
(
1
):
51
.
70.
Xu
S
,
Jiao
S
,
Guo
H
,
Chen
W
,
Yao
S
.
IMPeTUs parameters correlate with clinical features in newly diagnosed multiple myeloma
.
Eur J Radiol Open
.
2024
;
13
:
100598
.
71.
Davies
FE
,
Rosenthal
A
,
Rasche
L
, et al
.
Treatment to suppression of focal lesions on positron emission tomography-computed tomography is a therapeutic goal in newly diagnosed multiple myeloma
.
Haematologica
.
2018
;
103
(
6
):
1047
-
1053
.
72.
Charalampous
C
,
Goel
U
,
Broski
SM
, et al
.
Utility of PET/CT in assessing early treatment response in patients with newly diagnosed multiple myeloma
.
Blood Adv
.
2022
;
6
(
9
):
2763
-
2772
.
73.
Khan
MA
,
Markabawi
D
,
Alberti
M
, et al
.
Implications of 18F-FDG PET/CT findings before autologous stem cell transplant in patients with multiple myeloma
.
J Clin Oncol
.
2022
;
40
(
suppl 16
):
e20023
.
74.
Bailly
C
,
Carlier
T
,
Jamet
B
, et al
.
Interim PET analysis in first-line therapy of multiple myeloma: prognostic value of ΔSUVmax in the FDG-avid patients of the IMAJEM study
.
Clin Cancer Res
.
2018
;
24
(
21
):
5219
-
5224
.
75.
Nanni
C
,
Zamagni
E
,
Celli
M
, et al
.
The value of 18F-FDG PET/CT after autologous stem cell transplantation (ASCT) in patients affected by multiple myeloma (MM): experience with 77 patients
.
Clin Nucl Med
.
2013
;
38
(
2
):
e74
-
e79
.
76.
Kaddoura
M
,
Dingli
D
,
Buadi
FK
, et al
.
Prognostic impact of posttransplant FDG PET/CT scan in multiple myeloma
.
Blood Adv
.
2021
;
5
(
13
):
2753
-
2759
.
77.
Nørgaard
JN
,
Abildgaard
N
,
Lysén
A
;
Nordic Myeloma Study Group
.
Intensifying treatment in PET-positive multiple myeloma patients after upfront autologous stem cell transplantation
.
Leukemia
.
2023
;
37
(
10
):
2107
-
2114
.
78.
Schroeder
T
,
Martens
T
,
Chitadze
G
, et al
.
Pseudoprogression as adverse event of chimeric antigen receptor T cell therapy
.
Blood
.
2023
;
142
(
suppl 1
):
6935
.
79.
Jamet
B
,
Bodet-Milin
C
,
Moreau
P
,
Kraeber-Bodéré
F
,
Touzeau
C
.
2-[18F]FDG PET/CT flare-up phenomena following T-cell engager bispecific antibody in multiple myeloma
.
Clin Nucl Med
.
2023
;
48
(
5
):
e230
-
e231
.
80.
Zamagni
E
,
Nanni
C
,
Dozza
L
, et al
.
Standardization of 18F-FDG-PET/CT according to Deauville criteria for metabolic complete response definition in newly diagnosed multiple myeloma
.
J Clin Oncol
.
2021
;
39
(
2
):
116
-
125
.
81.
Zamagni
E
,
Oliva
S
,
Gay
F
, et al
.
Impact of minimal residual disease standardised assessment by FDG-PET/CT in transplant-eligible patients with newly diagnosed multiple myeloma enrolled in the imaging sub-study of the FORTE trial
.
EClinicalMedicine
.
2023
;
60
:
102017
.
82.
Messiou
C
,
Hillengass
J
,
Delorme
S
, 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
(
1
):
5
-
13
.
83.
Moreau
P
,
Zweegman
S
,
Perrot
A
, et al
.
Evaluation of the prognostic value of positron emission tomography-computed tomography (PET-CT) at diagnosis and follow-up in transplant-eligible newly diagnosed multiple myeloma (TE NDMM) patients treated in the phase 3 Cassiopeia study: results of the Cassiopet companion study
.
Blood
.
2019
;
134
(
suppl 1
):
692
.
84.
Rasche
L
,
Alapat
D
,
Kumar
M
, et al
.
Combination of flow cytometry and functional imaging for monitoring of residual disease in myeloma
.
Leukemia
.
2019
;
33
(
7
):
1713
-
1722
.
85.
Alonso
R
,
Cedena
MT
,
Gómez-Grande
A
, et al
.
Imaging and bone marrow assessments improve minimal residual disease prediction in multiple myeloma
.
Am J Hematol
.
2019
;
94
(
8
):
853
-
861
.
86.
Fonseca
R
,
Arribas
M
,
Wiedmeier-Nutor
JE
, et al
.
Correction: integrated analysis of next generation sequencing minimal residual disease (MRD) and PET scan in transplant eligible myeloma patients
.
Blood Cancer J
.
2023
;
13
(
1
):
87
.
87.
Hajiyianni
M
,
Sachpekidis
C
,
Kopp-Schneider
A
, et al
.
MRD by flow cytometry and FDG-PET/CT for the post-induction response assessment in patients with multiple myeloma treated in the phase 3 GMMG HD7 study
.
Blood
.
2024
;
144
(
suppl 1
):
4701
.
88.
Bansal
R
. JCO.
2023
.
89.
Schinke
C
,
Rasche
L
,
Ashby
C
, et al
.
Sequential imaging with diffusion-weighted whole-body MRI (DW-MRI) and PET-CT identifies patients at high risk of relapse in multiple myeloma
.
Blood
.
2023
;
142
(
suppl 1
):
882
.
90.
Hillengass
J
,
Bäuerle
T
,
Bartl
R
, et al
.
Diffusion-weighted imaging for non-invasive and quantitative monitoring of bone marrow infiltration in patients with monoclonal plasma cell disease: a comparative study with histology
.
Br J Haematol
.
2011
;
153
(
6
):
721
-
728
.
91.
Almeida
SD
,
Santinha
J
,
Oliveira
FPM
, et al
.
Quantification of tumor burden in multiple myeloma by atlas-based semi-automatic segmentation of WB-DWI
.
Cancer Imaging
.
2020
;
20
(
1
):
6
.
92.
Dutoit
JC
,
Verstraete
KL
.
MRI in multiple myeloma: a pictorial review of diagnostic and post-treatment findings
.
Insights Imaging
.
2016
;
7
(
4
):
553
-
569
.
93.
Ormond Filho
AG
,
Carneiro
BC
,
Pastore
D
, et al
.
Whole-body imaging of multiple myeloma: diagnostic criteria
.
Radiographics
.
2019
;
39
(
4
):
1077
-
1097
.
94.
Rodríguez-Laval
V
,
Lumbreras-Fernández
B
,
Aguado-Bueno
B
,
Gómez-León
N
.
Imaging of multiple myeloma: present and future
.
J Clin Med
.
2024
;
13
(
1
):
264
.
95.
Latifoltojar
A
,
Hall-Craggs
M
,
Rabin
N
, et al
.
Whole body magnetic resonance imaging in newly diagnosed multiple myeloma: early changes in lesional signal fat fraction predict disease response
.
Br J Haematol
.
2017
;
176
(
2
):
222
-
233
.
96.
Takasu
M
,
Kondo
S
,
Akiyama
Y
, et al
.
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
(
2
):
e0229607
.
97.
Rata
M
,
Blackledge
M
,
Scurr
E
, et al
.
Implementation of whole-body MRI (MY-RADS) within the OPTIMUM/MUKnine multi-centre clinical trial for patients with myeloma
.
Insights Imaging
.
2022
;
13
(
1
):
123
.
98.
Belotti
A
,
Ribolla
R
,
Cancelli
V
, et al
.
Predictive role of diffusion-weighted whole-body MRI (DW-MRI) imaging response according to MY-RADS criteria after autologous stem cell transplantation in patients with multiple myeloma and combined evaluation with MRD assessment by flow cytometry
.
Cancer Med
.
2021
;
10
(
17
):
5859
-
5865
.
99.
Torkian
P
,
Mansoori
B
,
Hillengass
J
, et al
.
Diffusion-weighted imaging (DWI) in diagnosis, staging, and treatment response assessment of multiple myeloma: a systematic review and meta-analysis
.
Skeletal Radiol
.
2023
;
52
(
3
):
565
-
583
.
100.
Ippolito
D
,
Giandola
T
,
Maino
C
, et al
.
Whole body low dose computed tomography (WBLDCT) can be comparable to whole-body magnetic resonance imaging (WBMRI) in the assessment of multiple myeloma
.
Diagnostics (Basel)
.
2021
;
11
(
5
):
857
.
101.
Sachpekidis
C
,
Mosebach
J
,
Freitag
MT
, et al
.
Application of (18)F-FDG PET and diffusion weighted imaging (DWI) in multiple myeloma: comparison of functional imaging modalities
.
Am J Nucl Med Mol Imaging
.
2015
;
5
(
5
):
479
-
492
.
102.
Mesguich
C
,
Hulin
C
,
Latrabe
V
, et al
.
Prospective comparison of 18-FDG PET/CT and whole-body diffusion-weighted MRI in the assessment of multiple myeloma
.
Ann Hematol
.
2020
;
99
(
12
):
2869
-
2880
.
103.
Westerland
O
,
Amlani
A
,
Kelly-Morland
C
, et al;
Myeloma Imaging Research Group at Guy’s & St Thomas’ Hospital, London and King’s College London
.
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
;
48
(
8
):
2558
-
2565
.
104.
Messiou
C
,
Porta
N
,
Sharma
B
, et al
.
Prospective evaluation of whole-body MRI versus FDG PET/CT for lesion detection in participants with myeloma
.
Radiol Imaging Cancer
.
2021
;
3
(
5
):
e210048
.
105.
Talarico
M
,
De Cicco
G
,
Tacchetti
P
, et al
.
OA-14 prospective functional bone disease evaluation of newly diagnosed multiple myeloma with combined use of (18)F-FDG-PET/CT and whole-body diffusion weighted magnetic resonance
.
Clin Lymphoma Myeloma Leuk
.
2024
;
24
(
suppl 2
):
S10
.
106.
Mai
EK
,
Hielscher
T
,
Kloth
JK
, et al
.
A magnetic resonance imaging-based prognostic scoring system to predict outcome in transplant-eligible patients with multiple myeloma
.
Haematologica
.
2015
;
100
(
6
):
818
-
825
.
107.
Rasche
L
,
Angtuaco
EJ
,
Alpe
TL
, et al
.
The presence of large focal lesions is a strong independent prognostic factor in multiple myeloma
.
Blood
.
2018
;
132
(
1
):
59
-
66
.
108.
Terao
T
,
Machida
Y
,
Narita
K
, et al
.
Total diffusion volume in MRI vs. total lesion glycolysis in PET/CT for tumor volume evaluation of multiple myeloma
.
Eur Radiol
.
2021
;
31
(
8
):
6136
-
6144
.
109.
Costachescu
D
,
Ionita
I
,
Borsi
EC
, et al
.
Whole-body diffusion-weighted magnetic resonance imaging and apparent diffusion coefficient values as prognostic factors in multiple myeloma
.
Exp Ther Med
.
2021
;
22
(
2
):
827
.
110.
Zhang
B
,
Bian
B
,
Zhang
Y
,
Zhang
L
,
Zhang
R
,
Wang
J
.
The apparent diffusion coefficient of diffusion-weighted whole-body magnetic resonance imaging affects the survival of multiple myeloma independently
.
Front Oncol
.
2022
;
12
:
780078
.
111.
Hillengass
J
,
Ayyaz
S
,
Kilk
K
, et al
.
Changes in magnetic resonance imaging before and after autologous stem cell transplantation correlate with response and survival in multiple myeloma
.
Haematologica
.
2012
;
97
(
11
):
1757
-
1760
.
112.
Giles
SL
,
Messiou
C
,
Collins
DJ
, et al
.
Whole-body diffusion-weighted MR imaging for assessment of treatment response in myeloma
.
Radiology
.
2014
;
271
(
3
):
785
-
794
.
113.
Wu
C
,
Huang
J
,
Xu
WB
, et al
.
Discriminating depth of response to therapy in multiple myeloma using whole-body diffusion-weighted MRI with apparent diffusion coefficient: preliminary results from a single-center study
.
Acad Radiol
.
2018
;
25
(
7
):
904
-
914
.
114.
Koutoulidis
V
,
Terpos
E
,
Papanikolaou
N
, et al
.
Comparison of MRI features of fat fraction and ADC for early treatment response assessment in participants with multiple myeloma
.
Radiology
.
2022
;
304
(
1
):
137
-
144
.
115.
Narita
K
,
Machida
Y
,
Ikeda
D
, et al
.
Total diffusion volume evaluated by whole-body diffusion weighted magnetic resonance image for the assessment of tumor burden in multiple myeloma; relations with measurable residual disease evaluated by next-generation flow cytometry and bone marrow CD138 staining assessed by Qupath
.
Blood
.
2024
;
144
(
suppl 1
):
491
.
116.
Rama
S
,
Suh
CH
,
Kim
KW
,
Durieux
JC
,
Ramaiya
NH
,
Tirumani
SH
.
Comparative performance of whole-body MRI and FDG PET/CT in evaluation of multiple myeloma treatment response: systematic review and meta-analysis
.
Am J Roentgenol
.
2022
;
218
(
4
):
602
-
613
.
117.
Belotti
A
,
Ribolla
R
,
Crippa
C
, et al
.
Predictive role of sustained imaging MRD negativity assessed by diffusion-weighted whole-body MRI in multiple myeloma
.
Am J Hematol
.
2023
;
98
(
9
):
E230
-
E232
.
118.
Belotti
A
,
Frittoli
B
,
Terlizzi
S
, et al
.
Early identification of functional high risk multiple myeloma patients after transplant with diffusion-weighted whole-body MRI: predictive role of fat fraction in addition to RAC score
.
Blood
.
2024
;
144
(
suppl 1
):
82
.
119.
Mesguich
C
,
Latrabe
V
,
Hulin
C
, et al
.
Prospective comparison of 18-FDG PET/CT and whole-body MRI with diffusion-weighted imaging in the evaluation of treatment response of multiple myeloma patients eligible for autologous stem cell transplant
.
Cancers (Basel)
.
2021
;
13
(
8
):
1938
.
120.
Böckle
D
,
Tabares
P
,
Zhou
X
, et al
.
Minimal residual disease and imaging-guided consolidation strategies in newly diagnosed and relapsed refractory multiple myeloma
.
Br J Haematol
.
2022
;
198
(
3
):
515
-
522
.
121.
Nosàs-Garcia
S
,
Moehler
T
,
Wasser
K
, et al
.
Dynamic contrast-enhanced MRI for assessing the disease activity of multiple myeloma: a comparative study with histology and clinical markers
.
J Magn Reson Imaging
.
2005
;
22
(
1
):
154
-
162
.
122.
Jamet
B
,
Necib
H
,
Carlier
T
, et al
.
DCE-MRI to distinguish all monoclonal plasma cell disease stages and correlation with diffusion-weighted MRI/PET-based biomarkers in a hybrid simultaneous whole body-2-[18F]FDG-PET/MRI imaging approach
.
Cancer Imaging
.
2024
;
24
(
1
):
93
.
123.
Hillengass
J
,
Ritsch
J
,
Merz
M
, et al
.
Increased microcirculation detected by dynamic contrast-enhanced magnetic resonance imaging is of prognostic significance in asymptomatic myeloma
.
Br J Haematol
.
2016
;
174
(
1
):
127
-
135
.
124.
Merz
M
,
Ritsch
J
,
Kunz
C
, et al
.
Dynamic contrast-enhanced magnetic resonance imaging for assessment of antiangiogenic treatment effects in multiple myeloma
.
Clin Cancer Res
.
2015
;
21
(
1
):
106
-
112
.
125.
Hillengass
J
,
Wasser
K
,
Delorme
S
, et al
.
Lumbar bone marrow microcirculation measurements from dynamic contrast-enhanced magnetic resonance imaging is a predictor of event-free survival in progressive multiple myeloma
.
Clin Cancer Res
.
2007
;
13
(
2 Pt 1
):
475
-
481
.
126.
Merz
M
,
Moehler
TM
,
Ritsch
J
, et al
.
Prognostic significance of increased bone marrow microcirculation in newly diagnosed multiple myeloma: results of a prospective DCE-MRI study
.
Eur Radiol
.
2016
;
26
(
5
):
1404
-
1411
.
127.
Dutoit
JC
,
Vanderkerken
MA
,
Verstraete
KL
.
Value of whole body MRI and dynamic contrast enhanced MRI in the diagnosis, follow-up and evaluation of disease activity and extent in multiple myeloma
.
Eur J Radiol
.
2013
;
82
(
9
):
1444
-
1452
.
128.
Dutoit
JC
,
Claus
E
,
Offner
F
,
Noens
L
,
Delanghe
J
,
Verstraete
KL
.
Combined evaluation of conventional MRI, dynamic contrast-enhanced MRI and diffusion weighted imaging for response evaluation of patients with multiple myeloma
.
Eur J Radiol
.
2016
;
85
(
2
):
373
-
382
.
129.
Lin
C
,
Luciani
A
,
Belhadj
K
, et al
.
Multiple myeloma treatment response assessment with whole-body dynamic contrast-enhanced MR imaging
.
Radiology
.
2010
;
254
(
2
):
521
-
531
.
130.
Bernstein
EJ
,
Schmidt-Lauber
C
,
Kay
J
.
Nephrogenic systemic fibrosis: a systemic fibrosing disease resulting from gadolinium exposure
.
Best Pract Res Clin Rheumatol
.
2012
;
26
(
4
):
489
-
503
.
131.
Burns
R
,
Mulé
S
,
Blanc-Durand
P
, et al
.
Optimization of whole-body 2-[18F]FDG-PET/MRI imaging protocol for the initial staging of patients with myeloma
.
Eur Radiol
.
2022
;
32
(
5
):
3085
-
3096
.
132.
Jamet
B
,
Carlier
T
,
Bailly
C
, et al
.
Hybrid simultaneous whole-body 2-[18F]FDG-PET/MRI imaging in newly diagnosed multiple myeloma: first diagnostic performance and clinical added value results
.
Eur Radiol
.
2023
;
33
(
9
):
6438
-
6447
.
133.
Barilà
G
,
Crimì
F
,
Arangio Febbo
M
, et al
.
Imaging minimal residual disease evaluation in multiple myeloma using [18F]FDG PET/MRI
.
Leuk Lymphoma
.
2023
;
64
(
2
):
503
-
506
.
134.
Chantry
A
,
Kazmi
M
,
Barrington
S
, et al;
British Society for Haematology Guidelines
.
Guidelines for the use of imaging in the management of patients with myeloma
.
Br J Haematol
.
2017
;
178
(
3
):
380
-
393
.
135.
Allegra
A
,
Tonacci
A
,
Sciaccotta
R
, et al
.
Machine learning and deep learning applications in multiple myeloma diagnosis, prognosis, and treatment selection
.
Cancers (Basel)
.
2022
;
14
(
3
):
606
.
136.
Jin
Z
,
Wang
Y
,
Wang
Y
,
Mao
Y
,
Zhang
F
,
Yu
J
.
Application of 18F-FDG PET-CT images based radiomics in identifying vertebral multiple myeloma and bone metastases
.
Front Med (Lausanne)
.
2022
;
9
:
874847
.
137.
Wennmann
M
,
Ming
W
,
Bauer
F
, et al
.
Prediction of bone marrow biopsy results from MRI in multiple myeloma patients using deep learning and radiomics
.
Invest Radiol
.
2023
;
58
(
10
):
754
-
765
.
138.
Alipour
E
,
Pooyan
A
,
Shomal Zadeh
F
,
Darbandi
AD
,
Bonaffini
PA
,
Chalian
M
.
Current status and future of artificial intelligence in MM imaging: a systematic review
.
Diagnostics (Basel)
.
2023
;
13
(
21
):
3372
.
139.
Piscopo
L
,
Scaglione
M
,
Klain
M
.
Artificial intelligence-based application in multiple myeloma
.
Eur J Nucl Med Mol Imaging
.
2024
;
51
(
7
):
1923
-
1925
.

Author notes

Individual participant data will not be shared.