Repeatability and Reproducibility of ADC Measurements and MRI Signal Intensity Measurements of Bone Marrow in Monoclonal Plasma Cell Disorders

2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Markus Wennmann ◽  
Heidi Thierjung ◽  
Fabian Bauer ◽  
Vivienn Weru ◽  
Thomas Hielscher ◽  
...  
2019 ◽  
Vol 2019 ◽  
pp. 1-6
Author(s):  
N. Steiner ◽  
R. Hajek ◽  
D. Nachbaur ◽  
B. Borjan ◽  
S. Sevcikova ◽  
...  

Introduction. The prognosis of multiple myeloma is still unfavorable due to inherent characteristics of the disease and the often-delayed diagnosis due to widespread and unspecific symptoms such as back pain and fatigue. Therefore, a simple diagnostic blood test would be helpful to speed up the diagnostic procedure in such patients (pts.). Here, we evaluated the diagnostic value of plasma levels of carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) in the peripheral blood and bone marrow of pts. with plasma cell disorders and in healthy controls. Materials and Methods. Immunoreactive CEACAM6 was determined in the peripheral blood and bone marrow (n=95/100) of pts. with monoclonal gammopathy of unknown significance (MGUS: 28/37), newly diagnosed multiple myeloma (NDMM: 42/40), and relapsed/refractory multiple myeloma (RRMM: 25/23) by sandwich ELISA. Results. Median CEACAM6 levels in the peripheral blood of pts. with plasma cell disorders were significantly higher than those of healthy controls (healthy controls: 15.2 pg/ml (12.1-17.1); MGUS: 19.0 pg/ml (16.4-22.5); NDMM: 18.0 pg/ml (13.4-21.2); and RRMM: 18.9 pg/ml (15.2-21.5); p<0.001). Plasma levels of CEACAM6 discriminated healthy subjects from MGUS/NDMM pts. (AUC=0.71, 95% CI: 0.6-0.8); i.e., a CEACAM6 level>17.3 pg/ml has an 82% (95% CI: 70-90) predictive probability for the identification of MGUS or NDMM. Moreover, CEACAM6 levels in the bone marrow were significantly higher in RRMM pts. than in NDMM pts. (p=0.04), suggesting a role of this molecule in disease progression. Conclusion. CEACAM6 plasma levels can noninvasively identify pts. with a plasma cell disorder and should be evaluated prospectively as a potential diagnostic marker. Moreover, due to high CEACAM6 levels in the bone marrow in RRMM pts., this adhesion molecule might be a therapeutic target in multiple myeloma pts.


2019 ◽  
Vol 19 (10) ◽  
pp. e333
Author(s):  
M Hasib Sidiqi ◽  
Mohammed Aljama ◽  
Shaji Kumar ◽  
Dragan Jevremovic ◽  
Francis Buadi ◽  
...  

Hemato ◽  
2021 ◽  
Vol 2 (4) ◽  
pp. 672-679
Author(s):  
Jens Hillengass ◽  
Maximilian Merz ◽  
Ronald Alberico ◽  
Majid Chalian

Multiple myeloma and other plasma cell disorders infiltrate the bone marrow in different patterns. While some patients show a homogeneous distribution of the clonal plasma cells others present with focal accumulations, commonly called focal lesions. Novel imaging techniques can provide information on these infiltration patterns and, due to their low invasiveness, can be performed repeatedly and therefore be used for monitoring. Conventional magnetic resonance imaging (MRI) has a high sensitivity for bone marrow assessment but cannot safely differentiate between active and inactive lesions. Therefore, positron emission tomography, especially combined with computed tomography (PET/CT), has been more widely used, at least for the monitoring of treatment response. Comparative, but mostly retrospective studies, have shown that functional MRI techniques, namely diffusion-weighted imaging (DWI), which assesses the movement of water molecules, can evaluate tissue cellularity with high sensitivity, which challenges the dominance of PET/CT in treatment response assessment. This review will discuss the benefits and challenges of DWI and compare it to other available imaging techniques used in patients with monoclonal plasma cell disorders.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5647-5647
Author(s):  
Neha Korde ◽  
Sham Mailankody ◽  
Evgeny Arons ◽  
Raul C. Braylan ◽  
Mark Raffeld ◽  
...  

Abstract Despite a reported increased incidence of secondary hematologic malignancies and plasma cell disorders in the literature, the specific co-existence of hairy cell leukemia (HCL) and monoclonal gammopathies of unknown significance (MGUS) or plasma cell multiple myeloma (MM) has been based on a few case reports. In the following report, we compile clinico-pathologic data on a series of hairy cell leukemia patients with myeloma precursor diseases using immunohistochemistry, molecular polymerase chain reaction (PCR) and multi-parametric flow cytometry (MFC) techniques with an aim to deep sequence these parallel lymphoid processes in the future. Between 2004 and 2014, 6 HCL patients followed at the National Institutes of Health were identified with associated plasma cell disorders, based on presence of increased clonal plasmacytosis in bone marrow and abnormal serum and/or urine protein electrophoresis/immunofixation. Immunohistochemical (IHC) staining for CD20, TRAP, CD138, cyclin D1, kappa and lambda light chains was performed on bone marrow biopsies. MFC using a panel of B-cell antibodies and PCR clonality studies targeting immunoglobulin heavy and light chain loci were performed prior to treatment to confirm the presence of malignant lymphoid clonal processes. Among the 6 hairy cell leukemia patients with associated plasma cell disorders, there were 5 males and one female. Median age was 67 (range, 47-74). Patients prior to treatment showed bone marrow involvement by HCL with 5-90% infiltration. Hairy cells in all patients tested were TRAP positive, Cyclin D1 was positive in 5, and BRAF V600E was detected in 5 of 5 patients tested. All patients were diagnosed as classic HCL, although 1 patient had an additional population consistent with HCL variant. In addition, 3/6 patients (50%) showed mild increase in marrow plasma cells (5-10%) and the other 3 patients (50%) had over 10% of plasma cells in the core biopsy. The mean plasma cell percentage was 10% (3-25%), mean monoclonal protein concentration was 1.3 g/dL, and isotypes included: 4 IgG, 1 IgA, and 1 free kappa only. 3 patients were classified as MGUS and 3 as smoldering MM (SMM). Interestingly, three out of 6 (50%) patients had positive cyclin D1 expression by IHC in both plasma cells and hairy cells. Several patients had evidence of multiple clonal rearrangements by PCR studies. In addition, two patients demonstrated evidence of monoclonal B-cell lymphocytosis (MBL) on MFC. So far, 3 patients achieved complete remission without minimal residual disease (MRD) using moxetumomab pasudotox in one multiply relapsed case, and first line cladribine plus rituximab in 2 newly diagnosed HCL cases, without significant progression to MM. No patients demonstrated end organ damage due to MGUS/SMM after a median follow-up of 4.6 (range 0.9-10.1) years. By using serum studies, IHC staining, PCR and MFC tools, we identified a group of HCL patients with evidence of additional precursor malignant lymphoid disease states, including MGUS/SMM and MBL. Underlying mechanisms of these parallel malignant processes may include global lymphoid dysregulation through common lymphocyte ancestry pathways; or, it could be due to post-immune HCL therapy exposure, or a combination. Currently, we are conducting deep sequencing of these samples with the aim to uncover mechanisms of pathogenesis. Treatments capable of eliminating HCL MRD, including addition of rituximab to first-line cladribine, or single-agent moxetumomab pasudotox for multiply relapsed HCL, might be advantageous for patients who may need treatment for MM in the future. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2014 ◽  
Vol 83 (6) ◽  
pp. 970-974 ◽  
Author(s):  
Mindaugas Andrulis ◽  
Tobias Bäuerle ◽  
Hartmut Goldschmidt ◽  
Stefan Delorme ◽  
Ola Landgren ◽  
...  

2014 ◽  
Vol 57 (2) ◽  
pp. 155-159 ◽  
Author(s):  
Julieta Panero ◽  
Nathan J. O’Callaghan ◽  
Michael Fenech ◽  
Irma Slavutsky

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5073-5073 ◽  
Author(s):  
Deborah Richardson ◽  
Elizabeth Hodges ◽  
Adnan Mani ◽  
Kim Orchard

Abstract CD66, a member of the carcinoembryonic antigen family, is known to be expressed on cells of myeloid and monocytic origin and has also been demonstrated on blasts from patients with B-lineage acute lymphoblastic leukaemia. An analysis of CD66 expression has been undertaken in bone marrow samples from patients with plasma cell disorders. Diagnostic bone marrow aspirate samples from 53 patients with multiple myeloma or monoclonal gammopathy were examined by multiparametric flow cytometry in order to demonstrate and quantitate plasma cells. Samples were analysed initially using a primary screening panel of antibodies including a combination of antiCD19 fluoroscein isothiocyanate (FITC)(Pharminogen), antiCD5 phycoerythrin (PE)(in-house), antiCD45 peridin chlorophyll protein (PerCP)(Becton Dickinson) and antiCD38 allophycocyanin (APC)(Phar). Samples shown to contain a CD45 negative, CD19 negative, CD38 positive population, consistent with the presence of plasma cells were then examined with a myeloma panel, including antiCD66. Samples from 41 patients were analysed using antiCD66 naked antibody (TheraPharm, GmBH) double-layered with sheep anti-mouse IgG F(ab’)2 fragment conjugated to FITC, antiCD138 PE, antiCD38 APC and antiCD45 PerCP. The correlation between CD38/CD138 and CD38/CD66 dual expression was 0.997. A further 13 samples were analysed using a commercial antiCD66 antibody conjugated to FITC (Dako). Again the correlation between CD38/CD138 and CD38/CD66 dual expression was 0.990. All patients apart from one, with morphologically detectable plasma cells, co-expressed CD66 with CD38. One patient with highly plasmablastic morphology did not express either CD38/138 or CD38/66 on plasma cells.The bone marrow of a normal individual was also examined and found to contain plasma cells which co-expressed CD19, CD138, CD38 and CD66. Plasma cells express CD66 in almost all patients with plasma cell disorders and expression has also been demonstrated on plasma cells in a normal individual. CD66 is therefore an attractive target for immunotherapy. A clinical trial using anti-CD66 targeted radiotherapy as part of the conditioning regimen for patients undergoing autologous or allogeneic stem cell transplantation, as therapy for multiple myeloma, is currently being conducted at our institution.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3603-3603
Author(s):  
Ruben A. Mesa ◽  
S. Vincent Rajkumar ◽  
Susan Schwager ◽  
Rebecca McClure ◽  
Heather Powell ◽  
...  

Abstract BACKGROUND: Bone marrow fibrosis might represent either a primary myeloid malignancy such as myelofibrosis with myeloid metaplasia (MMM) or a reactive phenomenon associated with other malignancies or inflammatory/infectious processes. The development of myelofibrosis in the context of a plasma cell dyscrasia is uncommon and incompletely understood from the standpoint of both pathogenesis and clinical implications. We sought to determine the origin and clinical ramifications of this phenomenon based on a consecutive series of patients with plasma cell disorders (PCD). METHODS: A retrospective search of institutional databases was performed for clinical cases of PCD with the concomitant observation of myelofibrosis. Bone marrow aspirates and trephines from the time of diagnosis of both PCD and myelofibrosis were reviewed. Clinical history was abstracted for clinical manifestations as well as disease course. DNA from archived cytogenetic pellets, when available, from the time of diagnosis, was screened for the presence of JAK2V617F. RESULTS: Among 7587 patients with a PCD seen over the last 30 years at our institution, 29 (0.3%) displayed concomitant myelofibrosis (median age 59 years, range 27–77; 52% males). The specific PCD was multiple myeloma in 25 patients, smoldering myeloma in 2, plasmacytoma in 1, and heavy chain disease in 1. Four patients (14%) had organomegaly and bone marrow histological features most consistent with MMM. Otherwise, myeloproliferative disorder (MPD)-characteristic features were infrequent and included leukocytosis and/or thrombocytosis in only 2 patients. None of the patients displayed thrombohemorrhagic complications. Archived DNA was available in 7 cases without either clinical or bone marrow histological evidence of a MPD and yet revealed the presence of JAK2V617F in 6 of the 7 cases (86%). In addition, one other patient without MPD phenotype displayed a monosomy 5 abnormality. To date, 2 patients, one with and one without a MPD phenotype underwent leukemic transformation. All other causes of death were related to PCD-associated complications. Median survival (Kaplan-Meier) for the patients with multiple myeloma was 50 months. Survival stratified by the myeloma international staging system (ISS) was similar or better than expected (published controls of 62, 44, and 29 months): median survivals for this series of 73, 51 and 37 months for stage I, II, and III disease, respectively. CONCLUSIONS: Myelofibrosis that accompanies a PCD often represents an underlying MPD and does not negatively impact the natural history of the associated PCD.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3098-3098 ◽  
Author(s):  
Brian A Walker ◽  
Dorota Rowczienio ◽  
Eileen M Boyle ◽  
Christopher P Wardell ◽  
Sajitha Sachchithanantham ◽  
...  

Abstract Systemic amyloid light chain amyloidosis (AL) is characterized by the deposition of immunoglobulin light chains as amyloid fibrils in different organs, where they form toxic protein aggregates. Most AL patients have relatively low levels of circulating free light chains and bone marrow plasmacytosis. The underlying disease is a plasma cell disorder, likely a monoclonal gammopathy, but limited data are available on the biology of the plasma cell clone underlying AL and existing studies have concentrated on chromosomal abnormalities. Many of the chromosomal abnormalities identified in AL are also seen in other plasma cell disorders, such as monoclonal gammopathy of undetermined significance (MGUS) and myeloma. These abnormalities include translocations involving the IGH locus, gains of 1q and deletions of 13q and 17p. Fluorescence in situhybridization studies have identified the translocation t(11;14) to be more frequent in AL and hyperdiploidy to be rare. The causal link between genetic changes in plasma cells and light chain instability remains unknown and progression to symptomatic myeloma is rare. We report the initial findings of the first exome sequencing to define the plasma cell signature in AL and compared this to MGUS and myeloma. CD138+ cells were selected using either EasySep (Stem Cell Technologies) or MACSort (Miltenyi) from the bone marrow of 18 AL patients and 5 MGUS patients. DNA was extracted from the CD138+ cells using the AllPrep kit (Qiagen). Non-involved DNA was isolated from peripheral white blood cells using the Flexigene kit (Qiagen). 200 ng DNA was subjected to exome sequencing using NEBNext kit (NEB) and SureSelect Human All Exon kit v5 and sequenced using 76-bp paired end reads. Fastq files were aligned to the reference genome using BWA and Stampy aligners. BAM files were recalibrated using the GATK and deduplicated using Picard. Paired tumour/normal BAMs were realigned together using the GATK indel realigner and SNVs were called using Mutect. Copy number data were estimated using the R package ExomeCNV. The median depth across all samples was 42x with 97% of the exome covered at 1x and 72% covered at 20x. Exome data to determine the cytogenetic groups of AL samples identified 42% hyperdiploid and 21% with t(11;14). The AL samples with t(11;14) did not contain any other copy number abnormalities. Exome sequencing on samples from patients with MGUS and myeloma was also performed to compare the genetic makeup and mutation spectrum of these well characterised plasma cell neoplasias with AL samples. MGUS samples had a median of 30 acquired nonsynonymous variants (range 24-189) and AL amyloidosis samples had a median of 17 acquired nonsynonymous variants (range 4-44). The AL samples had four recurrent mutations in PCMTD1 (n=3; L267F, P266S and M187I), C21orf33 (n=2; E72K), NLRP12 (n=2; L1018P, W959* ) and NRAS (n=2; Q61R, Q61H). In this small dataset, only 5 genes were mutated in both the MGUS and AL samples (DNMBP, FRG1, HIST1H1B, KRTAP4-11 and MCCC1). In order to assess the similarity (or differences) of plasma cells in AL to malignant plasma cells in general, we compared them to a random sampling of 20 multiple myeloma samples which had also been exome sequenced (median number of acquired nonsynonymous variants = 39 vs. 17 in AL samples). This revealed that the AL contained 21 mutated genes in common with the myeloma cohort, including DIS3 and NRAS. There were two DIS3 mutations in one AL sample at c.379D>E (p.D479E) and c.1999A>T (p.M667L), both of which were in the Ribonuclease II/R catalytic domain. Data on correlation of gene mutations and organ involvement in AL amyloidosis will be presented. We conclude that exome sequencing identifies a genetic signature of AL amyloidosis which is similar to other plasma cell disorders. This not only includes copy number abnormalities and translocations but also a similar number of nonsynonymous mutations to MGUS and fewer than the advanced myeloma samples. Study of further samples is in progress. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document