scholarly journals A Molecular Signature of Circulating MicroRNA Can Predict Osteolytic Bone Disease in Multiple Myeloma

Cancers ◽  
2021 ◽  
Vol 13 (15) ◽  
pp. 3877
Author(s):  
Aristea-Maria Papanota ◽  
Panagiotis Tsiakanikas ◽  
Christos K. Kontos ◽  
Panagiotis Malandrakis ◽  
Christine-Ivy Liacos ◽  
...  

Background: Multiple myeloma bone disease (MMBD) constitutes a common and severe complication of multiple myeloma (MM), impacting the quality of life and survival. We evaluated the clinical value of a panel of 19 miRNAs associated with osteoporosis in MMBD. Methods: miRNAs were isolated from the plasma of 62 newly diagnosed MM patients with or without MMBD. First-strand cDNA was synthesized, and relative quantification was performed using qPCR. Lastly, we carried out extensive biostatistical analysis. Results: Circulating levels of let-7b-5p, miR-143-3p, miR-17-5p, miR-214-3p, and miR-335-5p were significantly higher in the blood plasma of MM patients with MMBD compared to those without. Receiver operating characteristic curve and logistic regression analyses showed that these miRNAs could accurately predict MMBD. Furthermore, a standalone multi-miRNA–based logistic regression model exhibited the best predictive potential regarding MMBD. Two of those miRNAs also have a prognostic role in MM since survival analysis indicated that lower circulating levels of both let-7b-5p and miR-335-5p were associated with significantly worse progression-free survival, independently of the established prognostic factors. Conclusions: Our study proposes a miRNA signature to facilitate MMBD diagnosis, especially in ambiguous cases. Moreover, we provide evidence of the prognostic role of let-7b-5p and miR-335-5p as non-invasive prognostic biomarkers in MM.

2005 ◽  
Vol 65 (5) ◽  
pp. 1700-1709 ◽  
Author(s):  
Andrew C.W. Zannettino ◽  
Amanda N. Farrugia ◽  
Angela Kortesidis ◽  
Jim Manavis ◽  
L. Bik To ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5098-5098
Author(s):  
Melinda S. Gordon ◽  
Ariana M. Berenson ◽  
Charles B. Drucker ◽  
Matthew Katz ◽  
Hee Jin Lee ◽  
...  

Abstract Bone resorption leading to osteolytic bone disease is characteristic of multiple myeloma (MM). Recent studies show the presence of bone-resorbing osteoclasts and bone-forming osteoclasts in the circulation, and these cells may correlate with bone disease and change with anti-bone resorptive therapies. We have investigated whether there is an imbalance in the expression of osteoblast and osteoclast genes in the peripheral blood mononuclear cells (PBMCs) from MM patients relative to normal age-matched controls and the effect of bisphosphonate treatment on the expression of these genes. We analyzed the expression of a panel of osteoblast-related (bone alkaline phosphatase [bone AP], bone morphogenic protein 2 [BMP2], collagen I and osteocalcin) and osteoclast-related (b3 integrin, calcitonin, receptor for activation of nuclear factor kappa B [RANK] and tartrate-resistant alkaline phosphatase [TRAP]) genes by semi-quantitative RT-PCR on total RNA isolated from PBMCs obtained following density gradient separation. We demonstrated that the expression of the osteoblast-related gene BMP2 was reduced in eight of nine MM patients when compared with normal donors. In marked contrast, three osteoclast-related genes, b3 integrin, RANK and TRAP, were more highly expressed in all nine MM patients compared to the normal donors; only calcitonin expression was similar to the control subjects. Interestingly, patients receiving bisphosphonate treatment appeared to show increased osteoblast gene expression with higher amounts of bone AP, BMP2 and osteocalcin RNA compared to the patients not receiving anti-bone resorptive therapy. However, there was no alteration in the level of the RNA in any of the four osteoclast genes compared to patients not receiving anti-bone resorptive therapy. We are extending our analysis to a larger panel of MM patients in order to determine the relationship between these circulating cells and bone disease, overall clinical status and change in their levels with anti-bone resorptive therapy. In addition, we are also investigating whether there exist larger and smaller numbers of circulating osteoclasts and osteoblasts, respectively, in MM patients, or whether these circulating cells show alteration of their expression of these genes. Our semi-quantitative RT-PCR results are being correlated with immunohistochemical staining results from osteoblast and osteoclast markers obtained on PBMCs from MM and normal subjects. These studies provide evidence that the number of circulating osteoblasts and osteoclasts is altered in patients with MM, and also may suggest that bisphosphonate therapy may also be associated with changes in these cell populations.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1942-1942
Author(s):  
Houfu Leng ◽  
Adel Ersek ◽  
Emma Morris ◽  
Beatriz Gamez Molina ◽  
Claire M. Edwards ◽  
...  

Abstract Multiple myeloma (MM) is an incurable cancer of plasma cells (PC), with a median survival of 5-7 years. Osteolytic bone disease and skeletal complications occur in more than 80% of MM patients and significantly contribute to the morbidity and mortality of these patients. Glycosphingolipid (GSL), an essential constituent of the outer leaflet of the cellular membrane, is altered in MM and other hematological cancers. We previously reported that GM3, a subtype of GSL promotes osteoclastogenesis. On the other hand, the GSL synthase inhibitor N-butyl-deoxynojirimycin (NB-DNJ) reduces myeloma bone disease in the 5TGM1 mouse model of MM. Mechanistically, NB-DNJ prevents osteoclast (OC) development and activation by disrupting RANKL-induced localization of TRAF6 and c-SRC into lipid rafts and preventing nuclear accumulation of the transcriptional activator NFATc1. Although NB-DNJ is an FDA-approved drug treating Gaucher's disease, it has many undesired off-target effects, such as inhibiting lysosomal and plasma membrane Beta-glucocerebrosidase and interfering with intestinal glucosidases which leads to gastrointestinal toxicities and severe weight loss. Therefore, more specific GSL inhibitors are required to minimize the side effects. Here we report a novel GSL inhibitor called Genz112638 with comparable effects as NB-DNJ but reduced side effects. Genz112638 inhibits both OC formation (p < 0.01) and MM cell growth (p < 0.0001) in vitro in a dose-dependent manner. Moreover, compared to NB-DNJ, Genz112638 more significantly improved bone condition and potentially reduced MM burden, as evidenced by the amelioration of bone loss in the 5TGM1 model of myeloma, and a reduction in the proportion of MM within bone marrow and spleen without obvious adverse effects (n=6) (p < 0.01). As excessive malignant PC in MM normally arise from germinal centre, we also checked the effects of Genz112638 on germinal centre reactions in wildtype mice. We found that Genz112638 suppresses the formation of germinal centre B cells in mouse spleen induced by sheep red blood cells (n=7). Thus, Genz112638 may affect the pathogenesis of MM disease at the initial stage. Taken together, our data elucidate a novel specific GSL inhibitor as a promising candidate drug relieving two main features of MM: bone destruction and tumour burden with negligible side effects. In vitro, it decreases OC differentiation and proliferation, and meanwhile decreases MM viability and proliferation. In vivo, it may suppress B cell formation in germinal centre, ameliorate bone destruction, and potentially interfere with the vicious cycle between increased OC and susceptibility to MM. In short, we provide a preclinical platform for GSL inhibition as a new tool against MM and its related complications. Figure. Figure. Disclosures Horwood: Genzyme: Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3018-3018
Author(s):  
Roy Heusschen ◽  
Joséphine Muller ◽  
Marilène Binsfeld ◽  
Erwan Plougonven ◽  
Nadia Mahli ◽  
...  

Abstract Destructive bone lesions due to osteolytic bone disease are a major cause of morbidity and mortality in multiple myeloma patients, occurring in more than 80% of cases. Underlying osteolytic bone disease is an uncoupling of the bone remodeling process, with an increased activity of osteoclasts and a decreased activity of osteoblasts. Current strategies to treat osteolytic bone disease focus on anti-resorptive agents, which do not rebuild bone loss. Src kinase has been implicated in both osteoclast and osteoblast function. In this study, we assessed the effect of Src inhibition with AZD0530 (saracatinib, Astra Zeneca) on the development of multiple myeloma and its associated osteolytic bone disease. We first determined Src family kinase expression in the multiple myeloma microenvironment and found that patient-derived myeloma cells express Src at low levels but disease stage does not correlate with Src expression levels. In accordance with the literature, Src mRNA expression was found to increase during osteoclast differentiation and decrease during osteoblast differentiation in publicly available microarray datasets. Next, we validated an inhibitory role of AZD0530 on osteoclast differentiation and function. At a pharmacological relevant concentration of 1 micromolar, AZD0530 inhibited the differentiation of RAW264.7 osteoclasts (Oc.N/FOV: 15.5+-1.6 treated vs. 53+-1.5 non-treated). AZD0530 treatment appeared to hamper efficient progenitor cell fusion and osteoclast polarization, reflected by a decrease of CTSK and DC-STAMP mRNA levels and a defective actin ring formation in treated cultures, which culminated in a complete inhibition of bone resorption. When assessing the effect of AZD0530 on osteoblast function we found that AZD0530 inhibits osteoblast differentiation, with a decreased expression of OSX and OCN, and alters osteoblast morphology. In vivo, AZD0530 did not alter myeloma cell bone marrow infiltration in both the 5TGM.1 (37+-6.3% AZD0530 treated vs. 25.2+-6.7% non-treated) and 5T2MM (26.1+-7.7% vs. 29.1+-6.4%) murine multiple myeloma models. However, bone health was significantly improved in both models following treatment with AZD0530. In the 5TGM.1 model multiple trabecular bone parameters were restored to levels observed in healthy control mice following AZD0530 treatment, including BV/TV (11.7+-0.3% treated vs. 6.4+-0.3% non-treated), Tb.N. (2.5+-6x10^-2/mm vs. 1.7+-9x10^-2/mm) and Tb.Th (46.2+-1micron vs. 37+-0.8micron). These results were confirmed in the 5T2MM model, which displays a more severe osteolytic bone disease. In addition, AZD0530 treatment resulted in an increase in cortical thickness (157.8+-0.8micron treated vs. 151.4+-0.7micron non-treated) and a decrease in the number and size of cortical lesions in 5TGM.1 mice. Finally, our findings were corroborated by histomorphometric analyses. In conclusion, we report a potent inhibitory effect of the Src inhibitor AZD0530 on the development of osteolytic bone disease in multiple myeloma. Our results indicate that AZD0530 exerts this effect via the modulation of both osteoclast and osteoblast function. These findings warrant further study of the feasibility and efficacy of AZD0530 to treat osteolytic bone disease in multiple myeloma patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (23) ◽  
pp. 2656-2666
Author(s):  
Marita Westhrin ◽  
Vlado Kovcic ◽  
Zejian Zhang ◽  
Siv H. Moen ◽  
Tonje Marie Vikene Nedal ◽  
...  

Abstract Most patients with multiple myeloma develop a severe osteolytic bone disease. The myeloma cells secrete immunoglobulins, and the presence of monoclonal immunoglobulins in the patient’s sera is an important diagnostic criterion. Here, we show that immunoglobulins isolated from myeloma patients with bone disease promote osteoclast differentiation when added to human preosteoclasts in vitro, whereas immunoglobulins from patients without bone disease do not. This effect was primarily mediated by immune complexes or aggregates. The function and aggregation behavior of immunoglobulins are partly determined by differential glycosylation of the immunoglobulin-Fc part. Glycosylation analyses revealed that patients with bone disease had significantly less galactose on immunoglobulin G (IgG) compared with patients without bone disease and also less sialic acid on IgG compared with healthy persons. Importantly, we also observed a significant reduction of IgG sialylation in serum of patients upon onset of bone disease. In the 5TGM1 mouse myeloma model, we found decreased numbers of lesions and decreased CTX-1 levels, a marker for osteoclast activity, in mice treated with a sialic acid precursor, N-acetylmannosamine (ManNAc). ManNAc treatment increased IgG-Fc sialylation in the mice. Our data support that deglycosylated immunoglobulins promote bone loss in multiple myeloma and that altering IgG glycosylation may be a therapeutic strategy to reduce bone loss.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2503-2503
Author(s):  
Sonia Vallet ◽  
Noopur Raje ◽  
MariaTeresa Fulciniti ◽  
Kenji Ishitsuka ◽  
Teru Hideshima ◽  
...  

Abstract Osteolytic bone disease (OBD) is a frequent complication of multiple myeloma (MM), affecting 70 to 80% of the patients. OBD is characterized by imbalanced bone remodeling, due to decreased osteoblast (OB) number and increased osteoclast (OC) formation and activity. MM cells secrete osteoclastogenic factors, such as receptor activator of nuclear factor kappa B ligand (RANKL) and CCL3. In turn, OC support MM cell proliferation and survival, thus promoting a positive feedback that exacerbates bone resorption. Chemokines modulate osteoclastogenesis and promote MM cell proliferation, in particular CCL3 and its receptor CCR1 play an important role in mediating OBD in MM. MLN3897 (Millennium Pharmaceuticals, Cambridge) is a novel small molecule specific antagonist of human CCR1 (IC50 0.8 nM). It has a favorable toxicity profile in healthy volunteers and is currently undergoing phase II clinical trials in rheumatoid arthritis and multiple sclerosis. Here we evaluate the effects of MLN3897 on OC function and activity, as well as OC-MM cell interactions. Our in vitro data demonstrates a dual mechanism of action for MLN3897: it inhibits osteoclastogenesis and also overcomes the protective effects conferred by OC on MM cells. Our data further shows inhibition of OC formation and function by 40 and 70%, respectively, following MLN3897 treatment. This is mediated via inhibition of the fusion process and is accompanied by downregulation of pERK and c-fos signaling. To analyze its effect on MM cells, we verified CCR1 and CCR5 expression levels on MM1.S (15% and 3.6%) and OPM1 (3.8 and 0.7%). Our data show that OC secrete high levels of CCL3 which triggers MM cell migration; and that MLN3897 abrogates these effects by inhibiting the PI3K/Akt pathway. Moreover, MLN3897 overcomes the proliferative advantage conferred by OC on MM cells, as demonstrated in INA6, MM1.S and MM patient derived primary cells. OC induced MM cell proliferation is mediated by adhesion and cytokine secretion, and MLN3897 abrogates both MM cell-to-OC adhesion and interleukin-6 (IL6) secretion by OC in a co-culture system, thereby resulting in decreased MM cell survival and proliferation. To confirm these in vitro results, in vivo studies in a SCID-hu mouse model are underway. Implanted SCID-Hu INA-6 bearing mice are treated with twice daily oral MLN3897 for 3 weeks. The evaluation of osteolytic lesions and OC, OB and endothelial cell number; and tumor burden will be presented. Our in vitro results therefore show novel biologic sequelae of CCL3 and its inhibition on both osteoclastogenesis and MM cell growth. Our in vivo experiments will further validate the role of CCR1 in a human BM microenvironment-MM model, providing the framework for clinical trials of MLN3897 for the treatment of OBD in MM.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2963-2963 ◽  
Author(s):  
Evangelos Terpos ◽  
Dimitrios Christoulas ◽  
Maria Gkotzamanidou ◽  
Cornelia Bratengeier ◽  
Maria Gavriatopoulou ◽  
...  

Abstract Abstract 2963 Dickkopf-1 (Dkk-1) and sclerostin are inhibitors of the Wingless-type and integrase 1 (Wnt) signaling and they are implicated in the pathogenesis of multiple myeloma (MM) bone disease through inhibition of osteoblast function. There is very limited information for the circulating levels of Dkk-1 and sclerostin in different phases of MM and their alterations post therapies with novel agents. Therefore, we studied 284 MM patients (153M/131F, median age 66 years): 167 consecutive patients were newly-diagnosed (20 had asymptomatic MM and 147 symptomatic MM), 29 patients were at the plateau phase of MM and 88 patients had relapsed/refractory MM and received therapy with the combination of lenalidomide plus dexamethasone with or without bortezomib (VRD or RD; Dimopoulos et al, Leukemia 2010). For newly diagnosed patients, serum was stored at the time of diagnosis, while for patients at the plateau phase serum was collected at the time of confirmation of the plateau (at least 6 months with stable M-protein without criteria confirming progression) and for relapsed/refractory patients on day 1 of cycles 1, 4 and 7 of VRD or RD administration. Circulating levels of Dkk-1 and sclerostin were measured using ELISA methodology (R&D Systems, Minneapolis, MN, USA and Biomedica Medizinprodukte, Vienna, Austria, respectively) in all patients and in 20 gender- and age- matched healthy controls. Circulating Dkk-1 and sclerostin concentrations of newly diagnosed symptomatic patients (median: 1383 pg/mL, range:274-32, 862 pg/mL and 415 pg/mL, 0–3,340 pg/mL respectively) were increased compared to controls (1069 pg/mL, 540-2, 709 pg/mL; p<0.001 and 250 pg/mL, 0–720 pg/mL; p=0.03, respectively) and to asymptomatic patients at diagnosis (1044 pg/mL, 480-2, 335 pg/mL; p<0.001 and 140 pg/mL, 0–1,100 pg/mL; p=0.001, respectively). Patients at plateau phase had increased circulating levels of sclerostin (704 pg/mL, 68–2000 pg/mL; p <0.001) compared to controls (p=0.002) as well as to MM patients at diagnosis (p=0.02). In contrast, they had lower serum levels of Dkk-1 (1013 pg/mL, 414–1729 pg/mL) compared to MM patients at diagnosis (p<0.001) and no difference compared to controls. Patients with ISS-3 myeloma at diagnosis had higher values of Dkk-1 and sclerostin than ISS-1 and ISS-2 patients [median Dkk-1 values for ISS-1, ISS-2 and ISS-3 were: 1059 pg/mL, 1290 pg/mL and 2649 pg/mL, respectively; p(ANOVA)=0.031; median sclerostin values for ISS-1, ISS-2 and ISS-3 were: 394 pg/mL, 392 pg/mL and 714 pg/mL, respectively; p(ANOVA)=0.001]. Patients with lytic disease at diagnosis (n=116) had increased levels of Dkk-1 compared with patients with no lytic disease (n=51): 1475 pg/mL, 327-32, 862 pg/mL vs. 840 pg/mL, 274–1112 pg/mL; p=0.002. There was no difference in sclerostin levels between these patients; however, patients with advanced bone disease (>3 lytic lesions and/or a fracture) had a borderline increase in their circulating sclerostin compared to all others (p=0.072). Dkk-1 circulating levels correlated weakly with sclerostin (r=0.201, p=0.05). Relapsed patients had increased Dkk-1 (1218 pg/mL, 161-19, 325 pg/mL) and sclerostin (886 pg/mL, 90-6, 272 pg/mL) levels compared to controls and to asymptomatic patients at diagnosis (p<0.001 for all comparisons). In patients who received RD, Dkk-1 was increased and sclerostin was decreased after 6 cycles of therapy. Responders to RD had a median increase of 9% in Dkk-1 serum levels after 6 cycles of therapy, while non-responders had a median increase of 91% compared to baseline values (p<0.01). Patients who did not respond to RD showed an increase in bone resorption marker CTX (p=0.021) after 6 cycles of therapy. VRD administration resulted in a significant reduction of sRANKL (p=0.024) and increase of bone formation marker, osteocalcin (p=0.01) after 6 cycles, but showed only minimal reduction of Dkk-1 (p=0.08) and no alterations on sclerostin. In conclusion our study suggests that Dkk-1 is elevated in active myeloma, while sclerostin is elevated even in the plateau phase of the disease. Both correlated with adverse disease features. The increase of Dkk-1 by RD seems to be balanced by a reduction effect of bortezomib on Dkk-1 in VRD. Furthermore, the reduction of sclerostin in RD patients may represent a modulatory effect of lenalidomide on marrow microenvironment. These results further support the rationale for the use of drugs targeting Dkk-1 and sclerostin in MM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1920-1920
Author(s):  
Alessandra Romano ◽  
Antonella Chiechi ◽  
Calogero Vetro ◽  
Nunziatina Parrinello ◽  
Amy van Meter ◽  
...  

Abstract Abstract 1920 The biologic mechanisms involved in the pathogenesis of multiple myeloma (MM)-induced osteolytic bone disease are poorly understood. Physiological interactions between the serotoninergic and skeletal systems have been implicated by clinical observations. Brainstem-derived serotonin positively regulates bone mass following binding to 5-HT2C receptors on ventromedial hypothalamic neurons. This is opposed by platelet-derived serotonin that induces bone lysis and osteoclast activation. Moreover, the serotonin transporter SLCA6A4 is universally present among the malignant B cell clones. We examined serotonin dysregulation in two sample types from MM patients: peripheral blood and bone marrow. In the blood we measured by ELISA the ratio of serotonin in serum compared to platelets in patients with MM (n=10), MGUS (n=10) or healthy controls (n=5). We found higher levels of serotonin in platelets for the MM patients compared to the MGUS & healthy controls (p=0.017). Concomitantly there was less serotonin in the serum of MM patients compared to compared to the MGUS & healthy (p=0.002). This implies an imbalance in the compartmentalization of serotonin associated with presence of MM bone disease. Our multiplexed protein kinase signal pathway mapping technology, reverse phase protein microarrays (RPMA), was applied to bone marrow samples for quantifying post-translational modifications (e.g. phosphorylation, cleavage, acetylation) and/or total cell signaling kinase levels. Using reverse-phase protein microarray (RPMA) we retrospectively measured bone remodeling signal pathway perturbations in 15 bone marrow core biopsies from patients diagnosed with MM, at different clinical stages, and correlated this with the presence of MM-related bone disease (documented by scan or MRI). Bone marrow core biopsies exhibited significant elevation of cellular Serotonin, RANK, MMP-11, TNFα, TNF-R1, and Ezrin Tyr353 in MM with active bone disease (n=9) compared to patients without bone disease (n=6) (respectively p=0.031, p=0.038, p=0.0082, p=0.0221, p=0.01, p=0.028). To further evaluate the serotonin dysregulation in bone marrow cells, we measured serotonin bound to plasma cells (CD138+) compared to CD138- cells in bone marrow aspirates (n=21). Bone marrow aspirate were collected from patients undergoing standard of care hematological work up for multiple myeloma at any stage or treatment course. Patients with symptomatic myeloma (defined as presence of at least one of CRAB symptom) had lower serotonin levels in the CD138- bone microenvironment cells compared to non-symptomatic patients (p=0.0235). Plasma cells (CD138+) exhibited larger amounts of serotonin compared CD138- bone microenvironment cells (p=0.016). Taken together, our data show a dysregulation of serotonin in MM suggesting an altered distribution of serotonin in blood and bone marrow. This provides potential insights into diagnosis, prognosis, and/or treatment. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document