scholarly journals Defining the cell surface proteomic landscape of multiple myeloma reveals immunotherapeutic strategies and biomarkers of drug resistance

2021 ◽  
Author(s):  
Ian D. Ferguson ◽  
Bonell Patiño Escobar ◽  
Sami T. Tuomivaara ◽  
Yu-Hsiu T. Lin ◽  
Matthew A. Nix ◽  
...  

ABSTRACTThe myeloma cell surface proteome (“surfaceome”) not only determines tumor interaction with the microenvironment but serves as an emerging arena for therapeutic development. Here, we use glycoprotein capture proteomics to first define surface markers most-enriched on myeloma when compared to B-cell malignancy models, revealing unexpected biological signatures unique to malignant plasma cells. We next integrate our proteomic dataset with existing transcriptome databases, nominating CCR10 and TXNDC11 as possible monotherapeutic targets and CD48 as a promising co-target for increasing avidity of BCMA-directed cellular therapies. We further identify potential biomarkers of resistance to both proteasome inhibitors and lenalidomide including changes in CD53, EVI2B, CD10, and CD33. Comparison of short-term treatment with chronic resistance delineates large differences in surface proteome profile under each type of drug exposure. Finally, we develop a miniaturized version of the surface proteomics protocol and present the first surface proteomic profile of a primary myeloma patient plasma cell sample. Our dataset provides a unique resource to advance the biological, therapeutic, and diagnostic understanding of myeloma.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4450-4450
Author(s):  
Catherine M Gavile ◽  
David Egas ◽  
Gregory H Doho ◽  
Sagar Lonial ◽  
Kelvin P Lee ◽  
...  

Multiple myeloma is a malignancy of long lived plasma cells. Like normal plasma cells, myeloma cells are dependent on the bone marrow microenvironment for survival. While the specific interactions and downstream signals mediated by the bone marrow stroma have yet to be fully characterized, drug resistance is linked to these pathways, and further understanding will uncover new therapeutic avenues for myeloma. CD28 and CD86 are best known for their role in T-cell activation; however they have recently been shown to play important roles in the generation and survival of normal long lived plasma cells. CD28 is the canonical costimulatory receptor known to activate the PI3K-Akt pathway in T-cells upon binding to CD80 or CD86 from an antigen-presenting cell. CD28 and CD86 are also expressed by normal plasma and myeloma cells, and we have previously shown that both CD28 and CD86 are necessary for myeloma cell survival. Silencing of either CD28 or CD86 results in cell death in 3 human myeloma cell lines (RPMI8226, MM.1s, and KMS18). Interestingly, in 2 cell lines, knockdown of CD86 results in higher levels of cell death than CD28. In these lines, silencing CD28 or addition of a soluble inhibitor CTLA4-Ig (Abatacept), results in an increase in CD86 expression. Taken together, these data suggest that CD28 and CD86 regulate the survival of myeloma cells through either cis or trans signals, and feedback signals from CD28 control CD86 expression. The data also suggest that signaling events result from ligation of either CD28 or CD86. To better define the nature of the survival signals emanating from CD28 and CD86, we performed RNA-Seq on myeloma cells where CD28 or CD86 expression had been silenced. For silencing of CD28 we found 1292, 1195, and 1697 transcripts that were significantly changed compared to vector control in KMS18, MM.1s and RPMI8226, and silencing CD86 results in a similar number of changes (1405, 1200 and 1866 transcripts respectively). Since CD28 and CD86 form a receptor-ligand pair, we focused on genes that were common to silencing of both. Of genes that had significant expression changes compared to vector control, we found 229, 221 and 399 transcripts that were commonly regulated by CD28 and CD86 in KMS18, MM.1s and RPMI8226 respectively. Most transcripts were either upregulated (45.4 to 57.5%) or downregulated (28.8 to 41.5%) by silencing of either CD28 or CD86. A subset of transcripts (13.1 to 14%) showed a pattern of expression similar to CD86 - silencing of CD28 and CD86 had opposite effects on expression. This subset of transcripts may represent genes that are regulated by CD86 signaling, and may explain the difference in sensitivity to CD28 vs. CD86 silencing. Curiously, in KMS18 where this difference was not observed, RNA-Seq indicates that these cells are homozygous for a CD86 SNP that is associated with increased cancer susceptibility and lower transplant rejection, and may represent a hypomorphic allele. Surprisingly, we did not observe any significant changes in either pro- or anti-apoptotic Bcl-2 genes in any cell line except for upregulation in minor transcripts of Bcl2L11 (Bim) in KMS18 cells. This change did not affect overall expression as confirmed by qRT-PCR. However, expression of several cell surface proteins associated with myeloma cell survival did change. Integrin-ß1 (ITGB1) and -ß7 (ITGB7) are surface molecules that facilitate both cell-matrix and cell-cell interactions, and have been implicated in myeloma growth, survival, and drug-resistance. Knockdown of CD28 or CD86 resulted in downregulation of ITGB7 that was confirmed by qRT-PCR. We also saw a reduction of ITGB7 at the cell surface with CD86 knockdown, but not with CD28. ITGB1 expression was reduced at the mRNA and cell surface levels with knockdown of CD86, but was induced with CD28 knockdown. Based on their patterns of expression, ITGB7 may be regulated by CD28 signaling, while ITGB1 may be downstream of CD86 signaling. These data indicate that CD28-86 signaling regulates the expression of integrins on the surface of myeloma cells. Because drug resistance has been linked to the myeloma cells’ interaction with the bone marrow stroma and its resident cells (CAM-DR), these surface molecules could be important mediators of CD28 and CD86 survival signaling. Taken together, our data indicate that targeting CD28-86 signaling is a promising therapeutic approach to CAM-DR, and may be a useful addition to current regimens against myeloma. Disclosures: Lonial: Onyx: Consultancy; Celgene: Consultancy; Millennium: Consultancy; Novartis: Consultancy; BMS: Consultancy; Sanofi: Consultancy; Onyx: Consultancy. Boise:Onyx Pharmaceuticals: Consultancy.


2020 ◽  
Vol 3 ◽  
Author(s):  
Emily Adaniya ◽  
Christina Yu ◽  
Fabiana Perna

Background/Objective: Every year in the United States, 32,000 individuals are diagnosed with Multiple Myeloma (MM) and 13,000 die from the disease. MM is a cancer of the plasma cells and while treatment exists, MM remains incurable. Our overall goal is to identify biologically and therapeutically relevant cell surface targets to develop MM immunotherapy.      Methods: Given the genomic heterogeneity of MM, we performed surface analysis of seven MM cell lines by Mass-Spectrometry (MS), generating a pool of candidate proteins. Lacking specific tools for studying cell surface proteins, we developed an integrated computational tool with five distinct databases, and scored the likelihood of cell surface location for each molecule. One point was given for each database a protein appeared in. A protein receiving a score of three or more was considered a cell surface protein with high confidence. However, about one third of the candidates were not detected by the computational tool, thus requiring manual curation. We used UniProt and GeneCards to determine the subcellular locations of the unannotated IDs, and further confirmed their location with the computational tool.    Results: Of the 5,454 UniProt IDs produced by MS, 2,026 were not annotated. Causes of an unannotated ID included having an obsolete ID, being an isoform, and/or not being located on the cell surface. Through manual annotation, 8 unique cell surface IDs were added to the existing 448. The 456 targets were further analyzed based on their expression profiles in both MM and normal tissues resulting in 94 of the most promising targets, some of which would have been missed without manual curation.     Conclusion/Potential Impact: Public genomic databases are often incomplete and may contain errors making manual curation a necessary step in error reduction. Left unchecked, promising surface targets could have been overlooked hindering the creation of potentially curative immunotherapies. 


2021 ◽  
Vol 21 ◽  
pp. S94-S95
Author(s):  
Bonell Patino-Escobar ◽  
Corynn Kasap ◽  
Ian Ferguson ◽  
Martina Hale ◽  
Arun Wiita

2020 ◽  
Vol 8 (2) ◽  
pp. e000915
Author(s):  
Robyn A A Oldham ◽  
Mary L Faber ◽  
Theodore R Keppel ◽  
Amanda R Buchberger ◽  
Matthew Waas ◽  
...  

BackgroundMultiple myeloma (MM) is characterized by clonal expansion of malignant plasma cells in the bone marrow. While recent advances in treatment for MM have improved patient outcomes, the 5-year survival rate remains ~50%. A better understanding of the MM cell surface proteome could facilitate development of new directed therapies and assist in stratification and monitoring of patient outcomes.MethodsIn this study, we first used a mass spectrometry (MS)-based discovery-driven cell surface capture (CSC) approach to map the cell surface N-glycoproteome of MM cell lines. Next, we developed targeted MS assays, and applied these to cell lines and primary patient samples to refine the list of candidate tumor markers. Candidates of interest detected by MS on MM patient samples were further validated using flow cytometry (FCM).ResultsWe identified 696 MM cell surface N-glycoproteins by CSC, and developed 73 targeted MS detection assays. MS-based validation using primary specimens detected 30 proteins with significantly higher abundance in patient MM cells than controls. Nine of these proteins were identified as potential immunotherapeutic targets, including five that were validated by FCM, confirming their expression on the cell surface of primary MM patient cells.ConclusionsThis MM surface N-glycoproteome will be a valuable resource in the development of biomarkers and therapeutics. Further, we anticipate that our targeted MS assays will have clinical benefit for the diagnosis, stratification, and treatment of MM patients.


2021 ◽  
Author(s):  
Seiichi Okabe ◽  
Yuko Tanaka ◽  
Akihiko Gotoh

Abstract BackgroundMultiple myeloma (MM) is a hematological malignancy characterized by the clonal expansion of plasma cells in the bone marrow. The treatment of MM patients has been dramatically changed by new agents, such as proteasome inhibitors and immunomodulatory drugs; however, many patients will relapse, even if the new agents provide therapeutic advantages. Hypoxia is an important component of the bone-marrow microenvironment. 6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB) is responsible for maintaining cellular levels of fructose-2,6-bisphosphate, which regulates glycolysis.MethodsIn this study, we investigated the PFKFB functions in myeloma cells under hypoxic conditions. We also investigated whether PFKFB inhibitors could suppress myeloma cells and enhance their sensitivity to proteasome inhibition.ResultsWe first investigated the expression of PFKFBs in the myeloma cell lines under hypoxic conditions. Using public microarray datasets (GSE80140 and GSE80545), we found that the gene expressions of PFKFB3 and PFKFB4 were elevated under hypoxic conditions. Hypoxia-inducible factor 1α (HIF1α) was increased, and the phosphorylation of p38 mitogen-activated protein kinase (p38 MAPK) was activated. Under hypoxia, the activity of proteasome inhibitors was reduced. The PFKFB3 inhibitor, PFK158 and PFKFB4 inhibitor, 5MPN treatment were found to inhibit the growth of myeloma cells. The combined treatment of myeloma cells with carfilzomib and PFK158 or 5MPN was more cytotoxic than each drug alone. Caspase 3/7 activity and cellular cytotoxicity were also increased. In addition, we found that proteasomal activity was reduced by carfilzomib and PFK158 or 5MPN treatment. Intracellular adenosine triphosphate (ATP) levels drastically decreased after combined treatment. The combined treatment also changed the mitochondrial membrane potential in cell death and was effective on the bortezomib-resistant cell line.ConclusionPFKFB3 and PFKFB4 are enhanced in hypoxic conditions and are involved in proteasome-inhibitor sensitivity. Our data also suggested that administration of PFKFB3 and PFKFB4 inhibitors may be a powerful strategy against myeloma cells and may enhance the cytotoxic effects of proteasome inhibitors in hypoxic conditions.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 884-884
Author(s):  
Rakesh Bam ◽  
Wen Ling ◽  
Sharmin Khan ◽  
Angela Pennisi ◽  
Sathisha Upparahalli Venkateshaiah ◽  
...  

Abstract Abstract 884 Myeloma cells typically grow in bone, recruit osteoclast precursors and induce their differentiation and activity in areas adjacent to tumor foci. Clonal B-lymphocytes and plasma cells harboring lymphocytic phenotypes have a proposed role in sustaining myeloma. Bruton's tyrosine kinase (BTK) is expressed in hematopoietic cells and is particularly involved in B-lymphocyte function and osteoclastogenesis. The SDF-1/CXCR4 signaling pathway is reportedly involved in homing to bone of myeloma cells and osteoclast precursors. The aims of the study were to investigate the possible association between CXCR4 and BTK signaling in myeloma cells and osteoclast precursors, and the consequences of BTK inhibition on myeloma cell migration and clonogenicity, and osteoclastogenesis. By global gene expression profiling (GEP), BTK expression was moderately higher in clinical myeloma cells (n=559) than normal plasma cells (n=25, p<0.05). BTK was also detected in IL6–dependent cell lines and myeloma cell lines that can passage in vivo (n=7), using GEP, qRT-PCR and Western Blot analyses. Cell surface CXCR4 determined by flow cytometry was variably expressed in myeloma cells (5%–95%) and was significantly correlated with BTK gene expression (r=0.75, p<0.002, n=14). Most myeloma cell lines grown independently in vitro expressed very low levels of BTK; however, even in such cell lines (e.g. CAG), enrichment of myeloma cells expressing cell-surface CXCR4 by FACS analysis resulted in detectable BTK expression. BTK inhibition by shRNA in IL6–dependent INA6 cells inhibited their migration toward SDF-1 by >50% (p<0.006) and diminished their ability to form colonies in a 2-weeks clonogenic assay (p<0.0002). The BTK small molecule inhibitor LFM-A13 (25–50 μM) consistently reduced SDF-1-induced migration of primary myeloma cells and myeloma lines (n=6, p<0.03), and clonogenicity (colonies number and size) of 3 BTK-expressing myeloma lines by >50% (p<0.03). Using kinase immunoprecipitation assay, SDF-1 rapidly induced BTK phosphorylation (activation) in myeloma cells, an effect that was blocked by LFM-A13. In vivo, pretreatment of luciferase-expressing myeloma cells with LFM-A13 lessened their homing to bone following intravenous injection into SCID-rab mice as determined by live-animal imaging. BTK was highly expressed in osteoclast precursors while cell surface CXCR4 was detected in 5% of this cell population. LFM-A13 also reduced osteoclast precursor migration toward SDF-1 and suppressed osteoclast formation and bone resorption activity on dentine slices (p<0.002). In myeloma-bearing SCID-rab mice, LFM-A13 (n=10, 40 mg/kg, twice daily for 3 weeks, i.p.) reduced osteoclast number by 45% (p<0.004), prevented reduction of bone mineral density (BMD, p<0.04) and attenuated myeloma growth at near significant level (p<0.07). These data indicate that CXCR4 and BTK signaling are linked in myeloma cells and osteoclast precursors and that BTK has potential as a targeted myeloma therapy because of its roles in myeloma cell homing and clonogenicity and myeloma-induced osteolysis. Disclosures: Barlogie: Celgene, Genzyme, Novartis, Millennium: Consultancy, Honoraria, Patents & Royalties. Shaughnessy:Myeloma Health, Celgene, Genzyme, Novartis: Consultancy, Employment, Equity Ownership, Honoraria, Patents & Royalties.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3099-3099 ◽  
Author(s):  
Sathisha Upparahalli venkateshaiah ◽  
Rakesh Bam ◽  
Xin Li ◽  
Sharmin Khan ◽  
Wen Ling ◽  
...  

Abstract Identification of unique cell surface markers on myeloma cells is important for development of targeted therapies and detection of residual disease. GPRC5D is an orphan receptor reportedly expressed at high level in bone marrow (BM) aspirates or biopsies from myeloma patients. Other studies detected GPRC5D in skin and brain but not in other tissues. The aims of the study were to assess GPRC5D gene expression and cell surface localization in myeloma cells, the changes in expression of GPRC5D and other typical myeloma cell markers following coculture of primary myeloma cells with osteoclasts and the consequences of GPRC5D gene silencing on myeloma cell growth. Global gene expression profile (GEP), qRT-PCR and immunohistochemistry revealed exclusive high expression of GPRC5D in normal and myeloma plasma cells but not in B cells, MSCs, osteoclasts or BM mononucleated cells. GPRC5D expression was higher in myeloma plasma cells from newly diagnosed patients (n=698) compared to normal plasma cells (n=26, p<0.0001)). Among molecularly classified groups GPRC5D expression is higher in MS, MF and LB subgroups and lowest in CD2 and MM cell lines. Flow cytometry analysis and immunohistochemistry detected cell surface expression of GPRC5D in myeloma plasma cells while nuclear localization was also detected in certain myeloma cell lines (e.g. H929). Western blots analysis confirmed GPRC5D expression in whole cell lysate and nuclear fraction. We and other demonstrated phonotypical plasticity of myeloma plasma cells capable of altered expression of recognizable plasma cell markers (e.g. CD138, CD45) following coculture with stromal cells (Dezorella et al., 2009) or osteoclasts (Yaccoby, 2005). In coculture of primary CD138-selected myeloma cells with osteoclasts (n=8), CD138 (p<0,004), CD38 (p<0.001) and GPRC5D (p<0.007) were commonly and significantly downregulated, CD45 (p<0.02) was upregulated, and IRF4 expression was unaffected in cocultured myeloma cells compared to the control freshly obtained uncultured myeloma cells assessed by GEP and qRT-PCR. We also observed reduced expression of GPRC5D in MM cells purified from focal lesion compared to interstitial marrow in paired clinical samples (n=176, p<1.21E-09), suggesting that high activity of osteoclasts in osteolytic lesions mediates phenotypical alteration in myeloma cells. Stable knockdown of GPRC5D by 70% in CAG myeloma line using lentiviral particles containing shRNA had no effect on short-term growth of these cells assessed by MTT assay. These data indicate that GPRC5D is a novel cell surface marker for myeloma plasma cells and that its expression is reduced in dedifferentiated myeloma cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4715-4715
Author(s):  
Jason B. Brayer ◽  
Eva Sahakian ◽  
John Powers ◽  
Mark B Meads ◽  
Susan Deng ◽  
...  

Abstract While multiple myeloma (MM) remains incurable presently, expanded therapeutic options over the past decade have improved patient survival markedly. Proteasome inhibitors have redefined the treatment paradigm for myeloma, often serving as the backbone of front-line treatment. Histone deacetylase (HDAC) inhibitors (HDI), although only marginally active as single agent therapy in hematological malignancies, have demonstrated an ability to salvage bortezomib responsiveness in refractory patients, prompting heightened interest in this class of targeted therapeutics in myeloma. HDAC’s represent a family of enzymes, currently with 11 known members in the classical HDAC family, and subdivided into 4 sub-classes. HDAC11 is currently the only member of the sub-class IV and, as the newest member of the HDAC family, its impact on B cell lymphopoiesis and myeloma development is only starting to be unveiled. Intriguingly, we show that mice with germ-line silencing of HDAC11 (HDAC11KO mice) exhibit a 50% decrease in plasma cells in both the bone marrow and peripheral blood plasma cell compartments relative to wild-type mice. Consistent with this, Tg-HDAC11-eGFP mice, a transgenic strain engineered to express GFP under control of the HDAC11 promoter (Heinz, N Nat. Rev. Neuroscience 2001) reveals that HDAC11 expression is increased in the plasma cell population and to a lesser extent B1 B cells, as compared to earlier lineage stages. Similar observations based on measurements of HDAC11 mRNA were seen in normal human plasma cells. Significant increases in HDAC11 mRNA expression were observed in 7 of 11 primary human multiple myeloma samples and 11 of 12 human myeloma cell lines as compared to normal plasma cells, further emphasizing the potential relevance of HDAC11 to the underlying pathologic processes driving myeloma development and/or survival. Targeted silencing of HDAC11 in RPMI-8226 cells lines using siRNA results in a modest decrease in cell viability as measured by Annexin/PI staining and detection of activated caspase-3. Quisinostat, a second generation pan-HDI, has previously demonstrated activity against human myeloma cell lines in vitro (Stuhmer, Brit J Haematol, 2010), and suppressed bone destruction in an in vivo murine myeloma model (Deleu, Cancer Res, 2009). We similarly observe dose-dependent survival impairment in 10 human myeloma cell lines when cultured in the presence of quisinostat, with EC50’s consistently in the 1-10nM range. Importantly, quisinostat acts synergistically with proteasome inhibitiors (bortezomib and carfilzomib) in RPMI-8226 cells; more importantly, the degree of synergism is amplified in the RPMI-6226-B25 bortezomib-resistant cell line. Although a clear mechanism of action remains to be elucidated, preliminary data suggests that RPMI-8226 cells exposed to quisinostat appear to exhibit a decrease nuclear, but not cytosolic HDAC11. Collectively, these data illustrate a previously unknown role for HDAC11 in plasma cell differentiation and survival. Increased HDAC11 expression seen in myeloma patient specimens and primary myeloma cell lines highlights the potential of HDAC11 as a therapeutic target. Furthermore, we show that quisinostat, a pan-HDI with selectivity towards HDAC11 at lower dosing, acts synergistically with proteasome inhibitors in vitro in proteasome inhibitor sensitive and resistant cell lines. Future work will focus on further elucidating the role of HDAC11 in myeloma survival and drug response, with particular emphasis on proteasome inhibitors. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 2411
Author(s):  
Mélody Caillot ◽  
Hassan Dakik ◽  
Frédéric Mazurier ◽  
Brigitte Sola

Multiple myeloma (MM) is a common hematological disease characterized by the accumulation of clonal malignant plasma cells in the bone marrow. Over the past two decades, new therapeutic strategies have significantly improved the treatment outcome and patients survival. Nevertheless, most MM patients relapse underlying the need of new therapeutic approaches. Plasma cells are prone to produce large amounts of immunoglobulins causing the production of intracellular ROS. Although adapted to high level of ROS, MM cells die when exposed to drugs increasing ROS production either directly or by inhibiting antioxidant enzymes. In this review, we discuss the efficacy of ROS-generating drugs for inducing MM cell death and counteracting acquired drug resistance specifically toward proteasome inhibitors.


Hematology ◽  
2016 ◽  
Vol 2016 (1) ◽  
pp. 512-520 ◽  
Author(s):  
Hans C. Lee ◽  
Donna M. Weber

Abstract The use of proteasome inhibitors and immunomodulatory agents in the treatment of myeloma have resulted in significant improvements in patient outcomes over the last decade. Although these agents now form the backbone of current myeloma treatment regimens both in the frontline and in a relapsed setting, drug resistance remains an inevitable challenge that most patients will encounter during their disease course. Hence, new treatment strategies continue to be explored, and the recent regulatory approvals of the monoclonal antibodies (mAbs) daratumumab (DARA) and elotuzumab (ELO), which target the plasma cell surface proteins CD38 and signaling lymphocytic activation molecule F7 (SLAMF7), respectively, have heralded the long-awaited era of antibody-based approaches in the treatment of myeloma. Hoping to build on these advances, a number of other mAbs are in various stages of clinical development, including those targeting myeloma cell surface antigens, the bone marrow microenvironment, and immune effector T cells such as anti-programmed cell death protein 1 antibodies. In this review, the current landscape and practical use of mAb-based therapy in myeloma will be discussed.


Sign in / Sign up

Export Citation Format

Share Document