Comparison of Conventional Xrays with CT Based Approaches for Detection of Lytic Lesions in Multiple Myeloma

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 27-28
Author(s):  
Radhika Bansal ◽  
Sagar Rakshit ◽  
Katrina Glazebrook ◽  
Prashant Kapoor ◽  
Francis K. Buadi ◽  
...  

Background: Lytic bone lesions are one of the most common clinical characteristics of patients with multiple myeloma (MM) and identification of bone lesions help distinguish between patients with smoldering multiple MM and active MM. Given this, the most recent update of the diagnostic criteria for MM incorporates advanced imaging approaches for distinguishing between the two entities. Several small retrospective studies have compared conventional skeletal survey (SS) with whole-body low dose computed tomography (WBLDCT) scan or the CT portion of a positron emission tomography (PET) scan. Conducting prospective studies comparing these two modalities side by side is limited by the radiation exposure. We undertook this study to provide a comparison between these two imaging modalities in terms of their ability to recognize lytic bone lesions in patients with MM. Patients and methods: This was a retrospective study of consecutive patients with the diagnosis of MM treated at Mayo Cinic Hospital during 2004- 2018. Patients were included if they had a conventional skeletal survey no more than 3 months before MM diagnosis or any time after diagnosis and also had a WBLDCT or PET-CT within a 12-month period following the skeletal survey. We chose this approach since it is unlikely that a patient would have had both modalities done at the same time as part of standard of care (SOC). This approach was also facilitated by a gradual change in our SOC for skeletal imaging from conventional skeletal survey to WBLDCT. To measure the robustness of our findings, we performed a sensitivity analysis in patients who had the exam ≤6 months apart. Proportions were compared using a chi-square test and survival estimates were calculated using the Kaplan- Meier methodology and compared using log rank test. Results: The overall study cohort had 1040 patients, median age was 62 years at diagnosis (range, 24-94), 61% were male and 39% were female. The median time to the skeletal survey was 8 months from diagnosis (range, 0-160) and the median time to WBLDCT or PET-CT from SS was 2 months (0-12). A PET-CT was available in 789 (76%) of patients and WBLDCT in 251 (24%) of patients. Among the 300 patients with no lesions identified by SS, 188 (63%) had lytic lesions identified by CT, while in 60/740 (8%) patients with a positive SS did not have lytic lesions observed on the CT (p<0.0001). Overall, CT identified lytic lesions in 868 (84%) patients compared with 740 (71%) patients with lytic lesions seen on SS. Although the proportion of lytic lesions was slightly higher with PET compared to WBLDCT, the analysis did not reach statistical significance (65% vs. 56%; p = 0.17; Table 1a). After restricting the analysis to those with ≤6 months gap between low dose CT and SS (n=737), the result did not change (Table 1b). Further examination demonstrated that presence of lytic lesions by SS had no impact on OS from diagnosis, detection of lesions on the CT exam was associated with an inferior survival (112 vs. 81 months, p<0.0001). (Figure 1) Conclusion: Skeletal survey has been the screening technique of choice for evaluation of bone involvement in MM for decades. However, CT based approaches have higher sensitivity with lytic lesions identified in a higher proportion of patients. The prognostic value of lytic disease is evident with CT based detection, again suggesting that this provides a more accurate estimate of the skeletal disease burden. Low dose CT rather than conventional radiographs should be the modality of choice for monitoring disease-progression and diagnosing active multiple myeloma. Disclosures Kapoor: Cellectar: Consultancy; Janssen: Research Funding; Sanofi: Consultancy, Research Funding; Amgen: Research Funding; Takeda: Honoraria, Research Funding; GlaxoSmithKline: Research Funding; Celgene: Honoraria. Gertz:Abbvie: Other; Celgene: Other; Physicians Education Resource: Other: personal fee; Medscape: Other: personal fee, Speakers Bureau; Janssen: Other: personal fee; Spectrum: Other: personal fee, Research Funding; Johnson and Johnson: Speakers Bureau; Teva: Speakers Bureau; Annexon: Other: personal fee; Research to Practice: Other; Sanofi: Other; Proclara: Other; DAVA oncology: Speakers Bureau; Springer Publishing: Patents & Royalties; Appellis: Other: personal fee; Amgen: Other: personal fee; Prothena: Other: personal fee; Aurora Bio: Other; Alnylam: Other: personal fee; Ionis/Akcea: Other: personal fee. Dispenzieri:Takeda: Research Funding; Pfizer: Research Funding; Janssen: Research Funding; Intellia: Research Funding; Alnylam: Research Funding; Celgene: Research Funding. Dingli:Alexion: Consultancy; Janssen: Consultancy; Sanofi-Genzyme: Consultancy; Millenium: Consultancy; Bristol Myers Squibb: Research Funding; Rigel: Consultancy; Apellis: Consultancy; Karyopharm Therapeutics: Research Funding. Lin:Merck: Research Funding; Takeda: Research Funding; Legend BioTech: Consultancy; Juno: Consultancy; Bluebird Bio: Consultancy, Research Funding; Celgene: Consultancy, Research Funding; Novartis: Consultancy; Vineti: Consultancy; Janssen: Consultancy, Research Funding; Kite, a Gilead Company: Consultancy, Research Funding; Sorrento: Consultancy, Membership on an entity's Board of Directors or advisory committees; Gamida Cells: Consultancy. Kumar:Dr. Reddy's Laboratories: Honoraria; AbbVie: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Takeda: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Sanofi: Research Funding; Novartis: Research Funding; Janssen Oncology: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Genentech/Roche: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Merck: Consultancy, Research Funding; Adaptive Biotechnologies: Consultancy; Celgene/BMS: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Amgen: Consultancy, Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments, Research Funding; Carsgen: Other, Research Funding; Cellectar: Other; MedImmune: Research Funding; Karyopharm: Consultancy; BMS: Consultancy, Research Funding; Tenebio: Other, Research Funding; Genecentrix: Consultancy; Oncopeptides: Consultancy, Other: Independent Review Committee; IRC member; Kite Pharma: Consultancy, Research Funding.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 879-879
Author(s):  
Jing Fu ◽  
Shirong Li ◽  
Huihui Ma ◽  
G. David Roodman ◽  
Markus Y. Mapara ◽  
...  

Abstract Background Multiple myeloma (MM) cells secrete osteoclastogenic factors that activate osteoclasts (OCL) and contribute to development of pure lytic bone lesions in MM patients. We have recently shown that i) MMP13 is highly expressed by MM cells and ii) exogenous MMP13 increases OCL fusion and bone resorption (Feng et al, 2009). Further, MMP13 mediates these effects by upregulating dendritic cell-specific transmembrane protein (DC-STAMP), which is critical for OCL fusion and activation (Fu et al, 2012). Here, we investigated the role of MMP13 in MM-related bone disease (MMBD) in vivo and the underlying osteoclastogenic mechanisms. Methods and Results The role of MMP13 in MMBD was examined in vivo by the intratibial 5TGM1-GFP mouse MMBD model. Mouse MM cell line 5TGM1-GFP cells were transduced by pLKO.1-puro empty vector (EV) or sh-MMP13 (MMP13-KD) lentivirus followed by puromycin selection for 2 weeks. MMP13 knockdown in 5TGM1-MMP13-KD cells were confirmed by quantitative RT-PCR. 1×105 5TGM1-GFP-EV and 5TGM1-GFP-MMP13-KD cells were bilaterally intratibially injected into Recombination Activating Gene 2 (Rag2) knockout mice (n=9). After 4 weeks of tumor growth, tibiae were separated for micro quantitative computed tomography (micro-QCT) followed by immunohistochemistry (IHC) analysis. Following 5TGM1-GFP-EV injection, micro-QCT analysis of the tibiae and adjacent femurs indicated severe bone erosions, especially within trabecular bone. By contrast MMP13 KD inhibited the development of MM-induced bone lesions. Bone histomorphologic analysis showed that compared to 5TGM1-GFP-EV, MMP13-KD significantly reduced the MM induced trabecular bone loss with increased relative bone volume (0.069 ± 0.018 vs 0.0499 ± 0.016%; P=0.001), connective density (54.94 ± 33.03 vs 27.33 ± 18.97mm3; P=0.002), trabecular bone numbers (3.26 ± 0.29 vs 3.06 ± 0.33mm-1; P=0.032) and bone mineral density (159.1 ± 20.7 vs 134.2 ± 18.6mg/cm3; P=6E-04); as well as decreased triangulation bone surface to volume ratio (66.12 ± 6.67 vs 73.28 ± 10.07; P=0.017) and triangulation structure model index (3.05 ± 0.36 vs 3.42 ± 0.35 mm-1; P=0.002). In accordance with our finding that MMP13 induced OCL fusion, IHC results confirmed the presence of smaller TRAP+OCLs adjacent to the tumor in mice injected with 5TGM1-GFP-MMP13-KD cells compared with 5TGM1-GFP-EV cells. Although MMP13 knockdown showed no effects on 5TGM1-GFP cell growth in vitro, in vivo tumor progression represented by fluorescence imaging and sera immunoglobin 2G level (0.96 ± 0.12 vs 1.10 ± 0.11 mg/ml) was significantly inhibited (P=0.009 and 0.03 respectively), indicating MMP13 depletion in MM cells impaired OCL activation which, in turn, failed to support MM cell growth in bone marrow microenvironment as effectively in EV control group. In vitro studies demonstrated that MMP13 directly induced ERK1/2 phosphorylation in pre-osteoclasts. Consistent with a critical role for ERK1/2 phosphorylation in regulating OCL formation, U0126 (ERK1/2 inhibitor) blocked MMP13-induced ERK1/2 phosphorylation, ERK1/2-dependent DC-STAMP upregulation and MMP13-induced OCL fusion (P<0.01). Conclusion Our results demonstrate that silencing MMP13 expression in MM cells inhibits MM cell-induced OCL fusion and development of lytic bone lesions in vivo, indicating that MMP13 is essential for MM-induced bone diseases. Further, MMP13 upregulates DC-STAMP expression and OCL fusion via the activation of ERK1/2 signaling. Our data suggest that targeting MMP13 may represent a novel therapeutic approach for the treatment of MMBD. Disclosures: Roodman: Amgen: Membership on an entity’s Board of Directors or advisory committees; Lilly: Research Funding. Lentzsch:Celgene: Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1145-1145 ◽  
Author(s):  
Sara Bringhen ◽  
Valeria Magarotto ◽  
Anna Marina Liberati ◽  
Angelo Belotti ◽  
Alessandra Larocca ◽  
...  

Abstract Background: Survival rates of multiple myeloma (MM) patients (pts) has improved over the past few years, but patients inevitably relapse and become more resistant to subsequent treatments. Carfilzomib and Pomalidomide were both approved for the treatment of relapsed/refractory MM (RRMM). Combinations including a proteasome inhibitor (PI) plus an immunomodulator (IMiD), such as Bortezomib-Lenalidomide-Dexamethasone (VRD) or Carfilzomib-Lenalidomide-Dexamethasone (CRD), showed a very high response rate with an acceptable toxicity. Moreover, in the CHAMPION1 study (Berenson et al Blood 2016), the weekly infusion of Carfilzomib showed to be as effective as the twice schedule. In this phase I/II study we assessed for the first time weekly Carfilzomib plus Pomalidomide and low dose Dexamethasone (wKPd) for the treatment of RRMM. Here we report preliminary results. Methods: the primary objective of the phase I part of the trial was to determine the maximum tolerated dose (MTD) of wKPd combination. The primary objective of the phase II was to determine the rate of partial response (PR). Patients with RRMM, who received 1-3 prior lines of treatments and were refractory to Lenalidomide were eligible. Treatment consisted of 28-day cycles of oral Pomalidomide at fixed dose of 4 mg on days 1-21 (1 week off), oral or intravenous (iv) Dexamethasone 40 mg on days 1,8,15,22 and iv Carfilzomib at escalating doses on days 1,8,15. Escalation started at the dose of 36 mg/m2 (0 level) and used a standard 3+3 schema based on dose-limiting toxicities (DLTs) occurring in cycle 1. Treatment was continued until relapse or intolerance. Results: A total of 57 patients were enrolled in 6 Italian centers. Fifty-two patients could be evaluated for this analysis (5 patients did not complete the first cycle yet). The median age was 62 years with a median time from diagnosis of 4 years. 17/39 (44%) of patients were considered high risk according to cytogenetic abnormalities [at least one among t (4;14) t (14;16) and deletion chromosome 17 (del17) detected by FISH]. In the phase I of the trial 15 patients were enrolled. The first 3 patients at the dose level 0 of Carfilzomib did not experience any DLT. In the next cohort with Carfilzomib 20/45 mg/m2 a G3 hypertension and a sudden death occurred. According to the protocol, 3 more patients were enrolled at dose level 0: 1 patient experienced G3 atrial fibrillation, 2 patients ≥ G3 hypertension. Considering the serious adverse events (SAEs) occurred, the trial was temporary stopped to evaluate the benefit of continuing the study. All the DLTs were cardiologic and occurred in patients with a prior history of cardiac disease. As per protocol, they were evaluated with ECG and echocardiogram before the enrolment and were considered eligible for the study. The safety committee established new procedures for the evaluation of cardiac function of potentially eligible patients, including 24 h continuing pressure monitoring before the enrolment and serial measurement of blood pressure during and after Carfilzomib infusions. Six more patients were enrolled at dose level -1 (Carfilzomib 20/27 mg/m2) and none experienced a DLT. The MTD was established at dose level -1 with Carfilzomib 20/27 mg/m2, Pomalidomide 4 mg and Dexamethasone 40 mg. In the phase II portion of the trial, 42 patients were enrolled. Considering both phase I and II portions of the study, the most frequent drug related, grade ≥ 3 AEs were hematologic (65% of neutropenia and 13% of thrombocytopenia) and cardiologic (17%, mainly hypertension). We recorded only 4% of infection and ≥ G3 peripheral neuropathy. The overall response rate (ORR) of phase I/II portions was 58% (30/52) including 25% (13/52) of ≥ very good partial remission (VGPR). The ORR of high risk patients was 44% (7/16) including 19% (3/16) of ≥ VGPR. With a median follow-up of 10 months, median progression free survival (PFS) was 9.5 months and the median overall survival was not reached. Conclusions: This is the first phase I/II trial that combined weekly Carfilzomib with Pomalidomide and Dexamethasone. This combination was highly effective in RRMM. After a median follow-up of 10 months, wKRd showed a double median PFS in comparison with Pomalidomide-low dose dexamethasone (Sanmiguel et al Lancet Oncology 2013): 9.5 vs 4 months respectively, confirming the efficacy of combining a PI with an IMiD. An updated analysis will be presented at the meeting. Disclosures Bringhen: BMS: Honoraria; Celgene: Honoraria; Janssen-Cilag: Honoraria; Amgen: Other: ADVISORY BOARD; Mundipharma: Other: ADVISORY BOARD; Karyopharm: Other: ADVISORY BOARD. Larocca:Celgene: Honoraria; Janssen-Cilag: Honoraria; Bristol-Myers Squibb: Honoraria; Amgen: Honoraria. Gaidano:Karyopharm: Consultancy, Honoraria; Novartis: Consultancy, Honoraria, Speakers Bureau; Gilead: Consultancy, Honoraria, Speakers Bureau; Morphosys: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Speakers Bureau; Roche: Consultancy, Honoraria, Speakers Bureau. Oliva:Amgen: Honoraria; Celgene: Honoraria; Takeda: Honoraria. Sonneveld:Amgen: Consultancy, Honoraria, Research Funding; Takeda: Consultancy, Honoraria; Janssen: Consultancy, Honoraria, Research Funding; Celgene: Honoraria, Research Funding; Karyopharm: Consultancy, Honoraria, Research Funding. Palumbo:Janssen Cilag: Honoraria; Takeda: Employment, Honoraria. Boccadoro:Janssen: Honoraria, Research Funding; BMS: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Amgen: Honoraria, Research Funding; Abbivie: Honoraria; Mundipharma: Research Funding; SANOFI: Honoraria, Research Funding; CELGENE: Honoraria, Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 12-13
Author(s):  
Iuliana Vaxman ◽  
Alissa Visram ◽  
Shaji K. Kumar ◽  
Angela Dispenzieri ◽  
Francis K. Buadi ◽  
...  

Introduction Autologous stem cell transplantation (ASCT) has been used for treating multiple myeloma (MM) for over three decades and is generally reserved for patients younger than 65 years of age. The definition of transplant eligible is ill-defined and different centers have different policies to determine which patients are transplant eligible. Some centers have an age cut-off, others use clinical judgment, and some use various frailty scores (a scoring system based on comorbidities and physical and cognitive assessments) aiming to objectively assess transplant eligibility. There are limited data about outcomes in patients ≥ 75 years. Aim To report on outcomes of ASCT in a cohort of patients with MM aged 75 years or older. Methods Retrospective study of all consecutive MM patients aged ≥ 75 years that underwent ASCT at Mayo Clinic, Rochester, Minnesota. Stem cell transplantation at our center is routinely performed as an outpatient, with patients being hospitalized when deemed clinically necessary upon physician review. Results Between October 2005 and March 2020, 46 patients aged 75 years or older, received an ASCT at Mayo Clinic, Rochester. The median hematopoietic stem cell transplantation specific comorbidity index (HCT-CI) was 0 (range 0-6) with 8 patients having HCT-CI of 5 or 6. Median time from diagnosis to ASCT was 6.45 months (IQR 5.2-10.52) and 54% received reduced intensity conditioning with melphalan 140 mg/m2. All patients except one (that was treated with dexamethasone only) received induction with novel agents (listed in table 1) and 6 patients (13%) received doublet induction. All others received triplet induction. 46% of patients completed the ASCT without requiring hospitalization and 54% (n=25) of patients required hospitalization with a median duration of hospital admission of 9 days (IQR 5-13). Reasons for hospitalization included fever or infection (32%), cardiac arrhythmia (36%) and dehydration (32%). Overall response rate was 100% with a complete response seen in 57% of patients and 16 patients achieving MRD negative sCR. Median overall survival and progression free survival for the cohort were 82 months and 33 months, respectively. One patient died within 100 days of transplant representing a 2% 100-day mortality rate. Univariable cox regression model that evaluated the effect of gender, high risk cytogenetics, hemoglobin, renal function and melphalan dose did not detect any variable that was predictive of OS or PFS (Table 3). Conclusions ASCT is efficacious and can be safely delivered in the outpatient setting in carefully screened patients aged 75 or above. An arbitrary cutoff for age should not be used to exclude patients from ASCT, rather a careful assessment of "physiological age" including performance status and co-morbidities is required by an experienced treating team. Disclosures Kumar: Amgen: Consultancy, Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments, Research Funding; Cellectar: Other; Carsgen: Other, Research Funding; Dr. Reddy's Laboratories: Honoraria; Genentech/Roche: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Sanofi: Research Funding; Kite Pharma: Consultancy, Research Funding; Novartis: Research Funding; Celgene/BMS: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Merck: Consultancy, Research Funding; MedImmune: Research Funding; BMS: Consultancy, Research Funding; Tenebio: Other, Research Funding; Karyopharm: Consultancy; AbbVie: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Janssen Oncology: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Takeda: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Genecentrix: Consultancy; Oncopeptides: Consultancy, Other: Independent Review Committee; IRC member; Adaptive Biotechnologies: Consultancy. Dispenzieri:Pfizer: Research Funding; Janssen: Research Funding; Alnylam: Research Funding; Intellia: Research Funding; Takeda: Research Funding; Celgene: Research Funding. Dingli:Bristol Myers Squibb: Research Funding; Rigel: Consultancy; Janssen: Consultancy; Alexion: Consultancy; Karyopharm Therapeutics: Research Funding; Apellis: Consultancy; Sanofi-Genzyme: Consultancy; Millenium: Consultancy. Kapoor:Cellectar: Consultancy; Takeda: Honoraria, Research Funding; Celgene: Honoraria; Amgen: Research Funding; Sanofi: Consultancy, Research Funding; Janssen: Research Funding; GlaxoSmithKline: Research Funding. Gertz:Prothena: Other: personal fee; Medscape: Other: personal fee, Speakers Bureau; Appellis: Other: personal fee; Alnylam: Other: personal fee; Ionis/Akcea: Other: personal fee; Janssen: Other: personal fee; Research to Practice: Other; Sanofi: Other; Teva: Speakers Bureau; Johnson and Johnson: Speakers Bureau; DAVA oncology: Speakers Bureau; Proclara: Other; Springer Publishing: Patents & Royalties; Celgene: Other; Physicians Education Resource: Other: personal fee; Aurora Bio: Other; Amgen: Other: personal fee; Annexon: Other: personal fee; Spectrum: Other: personal fee, Research Funding; Abbvie: Other.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. e18574-e18574
Author(s):  
Muhammad Jawad Popalzai ◽  
Homam Alkaied ◽  
Maryah Mansoor ◽  
Arnold Brenner ◽  
Qun Dai

e18574 Background: Whole body skeletal x-ray is considered a gold standard for detecting bone lesions in patients with plasma cell disorders. PET/CT has been increasingly used but its role is yet to be defined. We conducted this study to compare the role of these two imaging modalities in evaluation of plasma cell disorders. Methods: This is single institution, retrospective study to evaluate the role of skeletal survey and PET/CT in patients with multiple myeloma, smoldering myeloma and MGUS. Patients’ records, imaging reports and subsequent management plan were reviewed and compared. Results: A total of 16 patients were reviewed. Among them, 11 patients had multiple myeloma, 2 had smoldering myeloma, and 3 had MGUS. 7/11 patients with multiple myeloma had concordant findings on skeletal survey and PET. 3 of these patients had negative skeletal surveys but had positive finding on PET/CT. PET/CT also identified plasmacytomas in 2 patients. In 2 patients with smoldering myeloma, both skeletal survey and PET/CT were negative. 2/3 patients with MGUS had lytic lesions on skeletal surveys which were not revealed by subsequent PET/CT’s. Both patients were observed without treatment and at 2 years follow up did not show disease progression. Conclusions: Our retrospective analysis showed that skeletal survey is still important for base-line evaluation of bone lesions in multiple myeloma and related monoclonal disorders. PET/CT is more sensitive for detection of bone lesions and can also detect extraosseous lesions such as plasmacytomas. Using tumor metabolic activity, PET/CT may improve diagnostic accuracy and is complementary to conventional skeletal survey. [Table: see text]


Author(s):  
Paolo Spinnato ◽  
Giacomo Filonzi ◽  
Alberto Conficoni ◽  
Giancarlo Facchini ◽  
Federico Ponti ◽  
...  

: Bone disease is the hallmark of multiple myeloma. Skeletal lesions are evaluated to establish the diagnosis, to choose the therapies and also to assess the response to treatments. Due to this, imaging procedures play a key-role in the management of multiple myeloma. For decades, conventional radiography has been the standard imaging modality. Subsequently, advances in the treatment of multiple myeloma have increased the need for accurate evaluation of skeletal disease. The introduction of new high performant imaging tools, such as whole-body low dose computed tomography, different types of magnetic resonance imaging studies, and 18F-fluorodeoxyglucose positron emission tomography, replaced conventional radiography. In this review we analyze the diagnostic potentials, indications of use, and applications of the imaging tools nowadays available. Whole body low-dose CT should be considered as the imaging modality of choice for the initial assessment of multiple myeloma lytic bone lesions. MRI is the gold-standard for detection of bone marrow involvement, while PET/CT is the preferred technique in assessment of response to therapy. Both MRI and PET/CT are able to provide prognostic information.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 2-3
Author(s):  
Neeraj Sharma ◽  
James B Smadbeck ◽  
Nadine Abdallah ◽  
Kathryn E. Pearce ◽  
Yan Asmann ◽  
...  

Background: Multiple myeloma (MM) is an incurable plasma cell malignancy and genetic abnormalities contribute to disease heterogeneity and outcome. Primary abnormalities, namely recurrent immunoglobulin (Ig) heavy chain translocations and hyperdiploidy, occur early in disease course. Secondary events, such as MYC abnormalities occur upon progression. Earlier studies showed MYC abnormalities detected by FISH or by capture sequencing were independently associated with poor outcome (Walker, et al., BCJ, 2014), while recent studies using WGS did not support this finding (excluding MYC/IGL) (Mikulasova, et al. Haematologica, 2020; Misund, et al. Leukemia, 2020). We hypothesize these discrepancies are due to differences in methods and sensitivities of detection of MYC abnormalities by FISH vs. WGS. Given that MYC abnormalities often display remarkable genomic heterogeneity with numerous gene partners, reduced detection of MYC abnormalities by FISH is not unexpected. This hypothesis is supported by lower frequencies of MYC abnormalities found by FISH (15%) vs. NGS (30-35%) consistent with ~50% false-negative rate of the MYC FISH probe (Smadbeck, et al. BCJ, 2019). To better understand the role of MYC in myeloma disease outcome, we compared the MYC abnormality subtype identified by FISH or NGS vs. MYC gene expression levels and overall survival. Methods: We performed a retrospective study of newly diagnosed MM patients seen at Mayo Clinic or enrolled in the MMRF CoMMpass trial. For Mayo cases, MYC FISH results (breakapart probe, Abbott) were obtained from the Mayo Clinic Genomics database (N=1342) and mate pair sequencing (MPseq) was performed on 140 cases. For CoMMpass cases, we obtained tumor long-insert whole genome sequencing (WGS), RNA sequencing (RNAseq) for gene expression and clinical outcome data. Overall survival (OS) was defined as time from diagnosis to death from any cause or to last follow up. Survival curves were estimated using Kaplan Meier and compared using the Log-Rank test. Statistical analyses performed using SPSS and JMP with significance determined when P &lt;0.05. Results: We first evaluated the impact of MYC abnormalities on OS when detected by FISH or NGS. In Mayo cases, OS was significantly shorter in patients with MYC abnormalities compared to patients without MYC abnormalities using FISH (5.3 vs. 8.0 years, P&lt;0.001, N=1342). In contrast, there was no significant difference in OS between patients with or without MYC or abnormalities using MPseq or WGS in both the Mayo and CoMMpass cohorts (Mayo: 6.4 vs. and 6.9 years P=0.78, N=140; CoMMpass: 4.9 vs. and 5.1 years P=0.74, N=546). Since FISH-detected MYC abnormalities were associated with poor outcome, we evaluated differences in the types of MYC abnormalities identified FISH and genome sequencing; 270 of 658 CoMMpass cases had a MYC abnormality and 12 abnormality subgroups were identified. In the Mayo cases, FISH preferentially detected translocations and complex abnormalities and missed insertions with flanking duplicating sequences or terminal tandem duplications (TTD) that occur telomeric to MYC. Since the level of MYC expression should be a consequence of the various genomic abnormalities altering the MYC gene region, we compared MYC expression levels in relation to MYC abnormality subgroups. Highest expression was seen with MYC amplification, followed by Ig abnormalities, non-Ig abnormalities, complex deletion/duplications, proximal deletions, non-Ig insertions, terminal deletions, TTD, trisomy 8, no MYC structural variation, monosomy 8 and cases with MAX mutations had the lowest expression. Abnormalities identified by FISH had higher MYC expression (83.5 TPM) compared to cases predicted to be missed by FISH (63.2 TPM). We tested if high MYC expression, irrespective of MYC structural abnormality, was associated with differences in OS. Boxplot analysis was used to categorize MYC expression in 631 CoMMpass patients as top quartile/high MYC expression (Q4≥ 75 TPM, n=159) and bottom quartile/low MYC expression (Q1≤ 16.5 TPM, n= 158) (see Figure). OS was significantly shorter in patients with high MYC expression compared to patients with low MYC expression (4.6 vs. 5.3 years, P &lt;0.038). Conclusion: We show that FISH detects only a subset of the MYC abnormalities detected by genome sequencing, and that FISH-detected MYC abnormalities are associated with higher MYC gene expression and decreased survival. Figure 1 Disclosures Kumar: Kite Pharma: Consultancy, Research Funding; Janssen Oncology: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; AbbVie: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Oncopeptides: Consultancy, Other: Independent Review Committee; IRC member; Dr. Reddy's Laboratories: Honoraria; Cellectar: Other; Takeda: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Novartis: Research Funding; Tenebio: Other, Research Funding; Carsgen: Other, Research Funding; Amgen: Consultancy, Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments, Research Funding; Merck: Consultancy, Research Funding; Genecentrix: Consultancy; BMS: Consultancy, Research Funding; Karyopharm: Consultancy; Celgene/BMS: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Genentech/Roche: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Sanofi: Research Funding; MedImmune: Research Funding; Adaptive Biotechnologies: Consultancy.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1828-1828 ◽  
Author(s):  
Steven Schuster ◽  
Angela Dispenzieri ◽  
S. Vincent Rajkumar ◽  
Alvaro Moreno Aspitia ◽  
Robert Kyle ◽  
...  

Abstract Abstract 1828 Poster Board I-854 Background IgM Multiple Myeloma (MM) and Waldenstrom's Macroglobulinemia (WM) are two hematologic diagnoses with the common variable of an IgM monoclonal gammopathy. Distinguishing these two diagnoses is critical as the approach to therapy is different. A study by Avet-Loiseau et al demonstrated the presence of t(11;14) translocations in 7 of 8 patients with IgM MM that was absent in all of a series of 17 cases of WM (Semin Oncol 2003 30:2;153). However, 6 of the 8 IgM MM cases lacked classic lytic lesions. Method A priori, based on the literature and the natural history of MM, we defined IgM MM as a symptomatic clonal plasma cell proliferative disorder characterized by a serum IgM monoclonal protein (regardless of size) plus presence of t(11;14) on fluorescent in situ hybridization (FISH) and/or lytic bone lesions felt to be related to the underlying plasma cell disorder. The cases for the study were screened by a computerized database search for ‘IgM’ and ‘Myeloma’ of all patients seen at Mayo Clinic in the last 30 years at all three sites (Rochester, Arizona and Florida). Patients identified on the computerized screen were then audited by chart review to identify the study cohort. Results 38 cases were identified on initial screen of the computerized database as potential patients with IgM MM. Of these, a total of 22 cases met our specific definition of IgM myeloma (t(11;14 and/or lytic lesions). Of the remaining 16 cases, 8 were IgM MGUS, 5 were WM based on clinical presentation (hyperviscosity, lymphadenopathy and organomegaly) and biopsy findings of lymphoplasmacytic lymphoma, 1 was excluded due to lack of information, and the remaining 2 patients were indeed considered to have clinical IgM MM. Interestingly, these two patients did not have either the t(11;14) or lytic lesions, but rather had immunophenotypic features suggestive of MM and not WM (CD138+, CD20-). Table 1 summarizes the clinical characteristics of the 22 patients who met our criteria for IgM MM. All 22 patients had lytic bone lesions. Of the 17 evaluated with FISH, 6 (35%) demonstrated the t(11;14) abnormality. Median overall survival by Kaplan-Meier analysis was 37 months represented in Figure 1. Conclusion IgM MM is a discrete clinical entity that can and should be distinguished from WM. Our definition of IgM MM is designed to be specific and requires the presence of a symptomatic IgM secreting plasma cell proliferative disorder plus presence of t(11;14) and/or lytic bone lesions felt related to the underlying plasma cell disorder. In this, the largest series of patients with IgM MM, the t(11;14) abnormality is very specific for IgM MM, but may not be sensitive, being present in approximately 1/3 of patients. The median overall survival is similar to non-IgM myeloma patients treated during this period, and much shorter than what would be expected for WM. The minority of symptomatic patients with IgM monoclonal gammopathy who do not meet this criteria, but have immunophenotypic features more suggestive of MM rather than WM need further study. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1878-1878
Author(s):  
Adriana C Rossi ◽  
Tomer M Mark ◽  
Kevin Wood ◽  
Roger N Pearse ◽  
Faiza Zafar ◽  
...  

Abstract Background Conventional radiography remains the gold standard imaging modality for staging multiple myeloma (MM). Other imaging modalities have been evaluated in recent years, and been shown to provide additional information about disease burden and location. FDG-PET/CT has proven to be useful in the identification of extramedullary disease and in monitoring patients with non-secretory myeloma. In addition to diagnostic utility, FDG-PET/CT has also been shown to predict time to relapse in the setting of newly diagnosed MM. However, to our knowledge its utility as a prognostic indicator in relapsed or refractory disease has not been studied. Methods We conducted a retrospective analysis of 61 patients with relapsed or refractory multiple myeloma (RRMM) who underwent PET/CT imaging prior to receiving salvage chemotherapy on a therapeutic trial of ClaPD (clarithromycin, pomalidomide, dexamethasone). Patients were heavily pre-treated, having received a minimum of 3 prior lines of therapy (range 3-15). All imaging was performed on the same PET/CT system at a single institution. Each PET/CT was evaluated in blinded fashion by two independent nuclear medicine physicians, with attention to the number and type of lesion, maximum SUV, and presence or absence of extramedullary disease. Disease response evaluation was performed monthly, and measured according to the international uniform response criteria. Multivariate analysis was performed to assess relationships of the above variables to depth of response, progression free survival (PFS), and overall survival (OS). Results Of 61 evaluable patients, 23 (38%) had no lytic lesions, 12 (20%) had <5 lytic lesions, and 26 (42%) had >5 lytic lesions on FDG-PET/CT. It is worth noting that 10 patients (16%) were found to have extramedullary disease, 8 of whom had >5 lytic bone lesions. There was no correlation between FDG-PET/CT findings and depth of response or median PFS, however patients with >5 lytic lesions had a median OS of 5.8 months, while it has not yet been reached for the other groups. At a median follow up of 13.2 months, 17 patients (74%) with no lytic lesions and 7 (58%) of those with <5 lytic lesions are alive. Conclusions The presence of >5 lesions on PET/CT at time of relapse is associated with poor prognosis in our cohort of heavily pre-treated patients with relapsed or refractory multiple myeloma receiving salvage chemotherapy with ClaPD. The presence of extramedullary disease, seen mostly in patients with >5 lesions, may contribute to our findings. Further studies in patients with relapsed or refractory MM are needed to evaluate the prognostic utility of FDG-PET/CT in this setting, as well as to extend these findings to other salvage regimens. Disclosures: Rossi: Celgene: Speakers Bureau. Mark:Celgene: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding, Speakers Bureau; Millennium: Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau; Onyx: Research Funding, Speakers Bureau. Zafar:Celgene: Speakers Bureau; Millennium: Speakers Bureau; Onyx: Speakers Bureau. Pekle:Celgene: Speakers Bureau; Millennium: Speakers Bureau. Niesvizky:Millennium: The Takeda Oncology Company: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding, Speakers Bureau; Onyx: Consultancy, Honoraria, Research Funding, Speakers Bureau; Celgene: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding, Speakers Bureau.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1121-1121
Author(s):  
Martha L Louzada ◽  
Maria-Victoria Mateos ◽  
Lenicio Siqueira ◽  
Jose-Maria B Bermejo ◽  
Enrique M Ocio ◽  
...  

Abstract Background Malignancy is a well-recognized risk factor for venous thromboembolism (VTE). In multiple myeloma the incidence of VTE varies between 3% and 10%. Immunomodulatory drugs (IMiDs) play a crucial role in the treatment of myeloma and are known to be associated with an increased risk of arterial and venous thromboembolic events (TE). It appears that patients with newly diagnosed multiple myeloma (NDMM) are at higher risk for TE compared to patients with relapsed or refractory myeloma (RRMM) at the start of IMiD therapy. Lenalidomide is a second-generation IMiD which in combination to dexamethasone has shown to be an effective and well-tolerated therapy for patients with NDMM or RRMM. However, studies have consistently demonstrated the need for TE prophylaxis in patients receiving the combination lenalidomide-dexamethasone which leads to a 4.4-fold increased risk for VTE compared to dexamethasone alone in the absence of prophylactic anticoagulants. Panel consensus from the International Myeloma Work Group has agreed that the choice of thromboprophylaxis depends on the individual risk of TE, as determined by patient and treatment-related factors, such as obesity, prior VTE, central venous catheter, immobilization, recent surgery, comorbidities, use of erythropoietin stimulating agents and myeloma therapy. Aspirin (ASA) is recommended for patients with one or no risk factors, and LMWH for those with more than one risk factor. However, the optimal approach to thromboprophylaxis has not yet been established. In this study we sought to compare the efficacy of ASA or low molecular weight heparin (LMWH) or vitamin K Antagonists (VKA) in the prevention of VTE or arterial thromboembolism (ATE) in patients with myeloma using lenalidomide-based therapy. Methods We performed a retrospective chart review in 2 centres (London, Canada; and Salamanca, Spain) on patients with NDMM or RRMM multiple myeloma receiving lenalidomide-based therapy. We collected data from january 2010 to December2014. We included adult patients diagnosed with NDMM or RRMM receiving lenalidomide-based therapy. We did not include who received lenalidomide but refused or had contra-indication to thromboprophylaxis; or used single agent lenalidomide. Results We included 168 patients with multiple myeloma receiving lenalidomide-based therapy. 14 (8%) were NDMM and 154 (92%) had RRMM. Median age was 68 (31-89) and 106 (63%) were males. On average patients with RRMM had 1.6 previous treatments (range:1-11). 104 (62%) patients were low risk and 64 (38%) were high risk for TE. 140 (83%) patients received prophylaxis with ASA, 32 (19%) LMWH and 6 (4%) VKA. 10 patients started with prophylactic LMWH for an average of 2 months then were empirically switched over to ASA. In total, there were 18 (10.7%) TE of which, 16 (9.5%) were VTE: 3 PE, 12 DVT, 1 both. The relative risk for TE was the same regardless of risk stratification [RR=1.27 (95%CI 0.524 - 3.059; p=0.599)]. At TE diagnosis, 16 patients were on ASA, 1 on LMWH and 1 on VKA. The relative risk of TE was significantly higher for patients on ASA compared to LMWH or VKA [RR= 2.17 (0.522 - 9.03; p= 0.286). After the TE, all patients changed anticoagulation strategy: 15 of 16 (94%) patients with VTE switched to therapeutic LMWH and 1 who was on VKA had ASA added. In the 2 patients with ATE, 1 started on full dose LMWH and the other one continued on ASA. 16 of 18 patients with a TE continued on lenalidomide-based therapy. There was no recurrent arterial or venous TE within the first 6 months of anticoagulation after the TE. Univariate analysis suggested that BMI, use of ASA and sex could be potential predictors of TE; but the logistic regression was not statistically significant (Table). Conclusions In patients with multiple myeloma on lenalidomide-based therapy the preferred TE prophylactic approach is low dose ASA irrespective of patients' risk assessment for thromboembolism. However, VTE risk in these patients is not negligible (9.5%) and low dose ASA may not be the best prophylactic strategy for them. It appears that patients on ASA, obese and males are at higher risk for TE. Future studies are needed to confirm these assumpations. Table 1. Multivariate Analysis of the TE risk for patients with myeloma on lenalidomide-based therapy Odds Ratio Variable Point Estimate 95% CI p -value Sex 2.316 0.854 6.278 0.0989 BMI 1.705 0.501 5.804 0.3935 ASA 3.870 0.486 30.796 0.2010 Disclosures Louzada: Celegene: Consultancy, Other: advisory board and expert opinion; pfizer: Consultancy, Other: advisory board and expert opinion; janssen: Consultancy, Other: advisory board and expert opinion. Mateos:Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Consultancy, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Amgen: Consultancy, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Ocio:Array BioPharma: Consultancy, Research Funding; Celgene: Consultancy, Honoraria; Amgen/Onyx: Consultancy, Honoraria, Research Funding; Bristol Myers Squibb: Consultancy; Mundipharma: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; MSD: Research Funding; Pharmamar: Consultancy, Research Funding; Janssen: Honoraria. Porras:Celgene: Consultancy, Honoraria.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-15
Author(s):  
Abdullah S. Al Saleh ◽  
Alissa Visram ◽  
Harsh Parmar ◽  
Angela Dispenzieri ◽  
Eli Muchtar ◽  
...  

Introduction: In general, the use of an immunomodulator (IMiD), proteasome inhibitors (PI) and dexamethasone (dex) for the treatment of MM is associated with better outcomes. The management of elderly patients with multiple myeloma (MM) is challenging due to difficulty in managing their co-morbidities and inability to tolerate treatment side effects. We evaluated therapies and outcomes of elderly patients with newly diagnosed MM. Methods: This is a retrospective study of patients with MM who were &gt;75 years old treated at the Mayo Clinic, Rochester from January 2004 to January 2018. We included patients who were treated on clinical trials as well as off-trials. Patients were classified as receiving treatment with IMiD+PI+dex, alkylator+PI+steroid, IMiD+dex, PI+dex, alkylator+IMid+steroid, and other (alkylator with steroid or steroid only). Treatment response was documented as well as the progression-free (PFS), defined as the time from therapy initiation to disease relapse or death from any cause and overall survival (OS), defined as the time from start of treatment to death from any cause. A multivariate analysis for factors affecting OS was done including the following variables: being on a triplet combination (alkylator+PI+steroid, IMid+PI+dex, or alkylator +IMiD+steroid), revised international staging system (R-ISS)(stage 3 vs. 1-2), bone marrow plasma cell percentage (BMPC%)(&gt;60% vs. ≤60%), and receiving treatment during or after 2010 vs. before 2010. Analysis was done for patients treated off-trials, as well as, including trial patients. Results: We identified 394 patients with MM who were &gt;75 years old and 246 (62%) were male. For non-trial patients (n=350), IMiD+dex (32%) was the most commonly used regimen followed by alkylator with steroid or steroid only (20%), alkylator+PI+steroid (18%), and IMid+PI+dex (13%). The remaining patients were treated with PI+dex (12%) and alkylator +IMiD+steroid (5%). Forty-four patients (11%) were treated in clinical trials with alkylator+IMid+steroid (47%), IMiD+dex (25%), IMiD+PI+dex (14%), and alkylator+PI+steroid (14%). The median follow up was 45.9 months with an interquartile range of 28.2 to 75.6 months. Overall, achieving very good partial response or complete response was more likely in patients who were treated with an IMid+PI+dex (58%) or alkylator+PI+steroid (47%), compared to in other therapies (5-30%)(P&lt;0.0001). The PFS and OS for non-trial patients are displayed in Figure 1 (A,B) and for all, including trial patients in (C,D). Overall, the median OS was significantly longer in patients who were treated with a triplet in non-trial as well as all patients. In a multivariate for OS including non-trial patients, predictors for better OS included receiving a triplet (HR: 0.63, P=0.02) and not having an R-ISS stage 3 (HR: 0.39, P=0.001). This was also found when including trial patients (using a triplet, HR: 0.65, P=0.01 and not having an R-ISS stage 3, HR: 0.35, P=0.0002). Conclusion: In MM patients &gt;75 years old, being able to receive triplet therapy is associated with better survival. This study provides better understanding of the natural history of MM outside of trials in the elderly age group. Disclosures Dispenzieri: Celgene: Research Funding; Alnylam: Research Funding; Pfizer: Research Funding; Intellia: Research Funding; Takeda: Research Funding; Janssen: Research Funding. Dingli:Bristol Myers Squibb: Research Funding; Alexion: Consultancy; Millenium: Consultancy; Rigel: Consultancy; Sanofi-Genzyme: Consultancy; Apellis: Consultancy; Janssen: Consultancy; Karyopharm Therapeutics: Research Funding. Kapoor:GlaxoSmithKline: Research Funding; Amgen: Research Funding; Takeda: Honoraria, Research Funding; Sanofi: Consultancy, Research Funding; Janssen: Research Funding; Cellectar: Consultancy; Celgene: Honoraria. Gertz:Spectrum: Other: personal fee, Research Funding; Janssen: Other: personal fee; Prothena: Other: personal fee; Alnylam: Other: personal fee; Ionis/Akcea: Other: personal fee; Springer Publishing: Patents & Royalties; Proclara: Other; DAVA oncology: Speakers Bureau; Johnson and Johnson: Speakers Bureau; Teva: Speakers Bureau; Sanofi: Other; Research to Practice: Other; Celgene: Other; Abbvie: Other; Aurora Bio: Other; Physicians Education Resource: Other: personal fee; Medscape: Other: personal fee, Speakers Bureau; Amgen: Other: personal fee; Appellis: Other: personal fee; Annexon: Other: personal fee. Kumar:Carsgen: Other, Research Funding; Tenebio: Other, Research Funding; BMS: Consultancy, Research Funding; Karyopharm: Consultancy; MedImmune: Research Funding; Sanofi: Research Funding; Novartis: Research Funding; Kite Pharma: Consultancy, Research Funding; Oncopeptides: Consultancy, Other: Independent Review Committee; IRC member; Genentech/Roche: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Celgene/BMS: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Adaptive Biotechnologies: Consultancy; Merck: Consultancy, Research Funding; Amgen: Consultancy, Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments, Research Funding; Janssen Oncology: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Takeda: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; AbbVie: Other: Research funding for clinical trials to the institution, Consulting/Advisory Board participation with no personal payments; Dr. Reddy's Laboratories: Honoraria; Cellectar: Other; Genecentrix: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document