scholarly journals The Influence of the Bone Marrow Niche on Drug Response Phenotypes of Blood Cancers

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 262-262
Author(s):  
Sophie Rabe ◽  
Eva Schitter ◽  
Tobias Roider ◽  
Peter-Martin Bruch ◽  
Carolin Kolb ◽  
...  

Abstract Background: Signals provided by the microenvironment can modify and circumvent pathway activities that are therapeutically targeted by drugs. However, a systems-level understanding of how the microenvironment and the genetic and molecular alterations of the tumor interact with each other and contribute to drug resistance is lacking. Methods: To address this unmet need, we established an automated microscopy-based phenotyping platform that uses co-culture conditions mimicking the bone marrow environment. We cultured primary tumor cells from more than 100 leukemia patients (CLL, AML, MCL, T-PLL, HCL) with and without bone marrow stroma cell support in DMEM and 10% human serum and treated each condition with 57 drugs in 3 concentrations. After 72h of incubation, 22 000 images per patient were acquired and processed by our custom made image analysis pipeline. Our set-up allows us to increase sensitivity far beyond simple viability testing, as it reads out additional cell type specific features such as cell morphology, autophagy and cell-cell interactions. Results: Quality assessment revealed that in contrast to mono-culture conditions, assay plate edge effects can be avoided under stable stroma cell co-culture conditions. Correlation of replicated patient samples were comparable between mono- and co-cultures (R2>0.75). In the absence of their native microenvironment, primary leukemia cells undergo spontaneous apoptosis ex-vivo. Viability at culture start was always >90% and dropped to a median of 51% (viability range: 17%-90%) after 72h in mono-cultures. Among CLL samples spontaneous apoptosis was not dependent on either IGHV mutation status or any major cytogenetic risk group. Bone marrow stroma cell co-culture conditions protected tumor cells from spontaneous apoptosis (p=8.2e-6, paired t-test). Patient samples with a high degree of spontaneous apoptosis benefited most from co-culture conditions (p=7.2e-10, Pearson correlation). To model interactions of stroma cell conditions and drug-induced apoptosis we established the following linear model: Viability ~ drug-effect + culture-model + drug-effect:culture-model. While activity of some drugs was significantly altered under co-culture conditions, we could also identify drugs with similar activity in mono- and co-cultures. For instance, the activity of common chemotherapeutics (fludarabine: p=0.002 at 0.6µM, cytarabine: p=0.001 at 1.5µM, ANOVA) or bromodomain inhibitors (I-BET-762: p=5.9e-5 at 4.5µM, JQ1: p=1.5e-8 at 1.5µM, ANOVA) was significantly reduced under co-culture conditions. In contrast, PI3K inhibitors idelalisib and duvelisib had a similar activity in mono-culture and stroma co-culture conditions and might represent a starting point to overcome stroma cell mediated drug resistance. In CLL, we identified IGHV mutation status and trisomy 12 as important determinants of response to kinase inhibitors. We confirmed these findings in stroma cell co-cultures, e.g. a better activity of B-cell receptor inhibitors in trisomy 12 and IGHV unmutated CLL. A systematic comparison of ex-vivo drug response pattern in mono- and co-cultures across 171 drug conditions will be presented. Conclusion: Our results suggest that high throughput co-culture drug testing can be robustly performed and provide an unprecedented understanding of how the stroma cell microenvironment and the genetic make-up of tumor cells contribute to drug resistance and sensitivity. Figure: Over 2 million microscopy images were acquired and analysed to assess drug resistance and sensitivity in a co-culture model of primary leukemia and bone marrow stroma cells. blue= Hoechst33342, green=Calcein AM, red=lysosomal dye NIR Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.

2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Eimear O’ Reilly ◽  
Sukhraj Pal S. Dhami ◽  
Denis V. Baev ◽  
Csaba Ortutay ◽  
Anna Halpin-McCormick ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3288-3288
Author(s):  
Mark B Meads ◽  
Paula Oliveira ◽  
Allison Distler ◽  
Maria Silva ◽  
Karen Burger ◽  
...  

Abstract Multiple myeloma (MM) is a heterogeneous plasma cell neoplasm that remains all but incurable despite recent advances in treatment. Indeed, nearly all patients eventually experience disease progression or relapse due to a reservoir of residual myeloma cells that appear to persist through pro-survival signaling from interactions with the tumor microenvironment (TME), leading to eventual clonal expansion. Thus, identifying targets that are induced in MM by the TME may reveal new and important targets amenable to therapeutic intervention. To develop a non-biased method to screen bone marrow specimens from myeloma patients for activated targets throughout the course of disease, we used a combination of activity-based protein profiling (ABPP) and a high-throughput protein kinase inhibitor (PKI) screen using a platform that recapitulates the TME. Target validation was then performed using ex vivo functional screens of pathways using MM patient specimens. The MM cell lines MM1.S, H929, and OPM2 were grown in mono-culture or co-culture with HS5 bone marrow stroma cells for 24h and lysates were enriched for ATP binding proteins by affinity purification versus a chemical probe (ActivX, Thermo). Tryptic peptides were measured using discovery proteomics (nano-UPLC and QExactive Plus mass spectrometer). Using this method, 176, 136, and 85 kinases out of a total of 1511, 1409, and 1281 proteins were preferentially enriched by 2-fold change from MM1.S, H929, and OPM2 myeloma cells grown in co-culture conditions with HS5 bone marrow stroma, respectively. Of these, 42 kinases were common to all three and 87 were common to two of three MM cell lines. Kinases were chosen for target validation after pathway analysis using the Kyoto Encyclopedia of Genes and Genomes database to identify signaling networks. To identify functionally relevant signaling networks identified via ABPP experiments, the same MM cell lines were simultaneously screened with 30 protein kinase inhibitors (PKIs) in a novel high throughput viability assay. This label-free method measures the viability of MM cells grown in a collagen matrix with bone marrow stroma cells in 384-well plates to simulate the TME by capturing brightfield images every 30 minutes for 96h using a motorized microscope equipped with an incubation chamber. Digital image analysis software measures live cell numbers by detecting membrane motion and generates viability curves as a function of drug concentration and exposure time (Khin et al. Cancer Research 2014). This functional screen confirming known MM survival networks, validated 12 kinases/PKIs in the context of the TME and highlighted novel targetable pathways. To provide an additional level of screening, the same PKIs were tested in CD138-MACS-selected cells from 15 MM patient specimens in a high-throughput viability assay. Eight PKIs targeting IGFR, PLK1, Abl, mTOR, FAK/Pyk2, ALK, Akt, and Casein Kinase-1δ (CK1δ)/CK1ε also showed significant activity in the 15 primary MM specimens. Our three-tiered pharmaco-proteomic screen identified eight kinases critical to MM survival in the context of the TME. Notably, a highly specific in-house inhibitor of Casein Kinase 1δ/CK1ε, SR-3029, which targets the Wnt/β-catnenin pathway, was identified as the most effective compound assessed as a single agent in our ex vivo viability assay in all patients with an average 36h LD50 of 290nM. This compound is under further investigation in MM (Submitted Abstract: Burger, et al, ASH 2016). Additional studies are underway to functionally interrogate the pathways identified in this screen, including ErbB1/EGFR, EphA1 and AMPK. Future work will optimize this method for evaluation of primary bone marrow specimens with ABPP followed by functional validation to better predict potential clinical response at different disease stages. We anticipate that this iterative "at the moment of care" approach is critical because drug resistant tumor phenotypes fluctuate with therapy, and this strategy can track and define clinically relevant changes in tumor cells in situ after the selection pressures applied by exposure to therapy. Disclosures Shain: Novartis: Speakers Bureau; Amgen/Onyx: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Takeda/Millennium: Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Signal Genetics: Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau.


Blood ◽  
2010 ◽  
Vol 115 (14) ◽  
pp. 2827-2834 ◽  
Author(s):  
James J. Driscoll ◽  
Dheeraj Pelluru ◽  
Konstantinos Lefkimmiatis ◽  
Mariateresa Fulciniti ◽  
Rao H. Prabhala ◽  
...  

Abstract Multiple myeloma (MM) is a plasma cell neoplasm that proceeds through a premalignant state of monoclonal gammopathy of unknown significance; however, the molecular events responsible for myelomagenesis remain uncharacterized. To identify cellular pathways deregulated in MM, we addressed that sumoylation is homologous to ubiquitination and results in the attachment of the ubiquitin-like protein Sumo onto target proteins. Sumoylation was markedly enhanced in MM patient lysates compared with normal plasma cells and expression profiling indicated a relative induction of sumoylation pathway genes. The Sumo-conjugating enzyme Ube2I, the Sumo-ligase PIAS1, and the Sumo-inducer ARF were elevated in MM patient samples and cell lines. Survival correlated with expression because 80% of patients with low UBE2I and PIAS1 were living 6 years after transplantation, whereas only 45% of patients with high expression survived 6 years. UBE2I encodes the sole Sumo-conjugating enzyme in mammalian cells and cells transfected with a dominant-negative sumoylation-deficient UBE2I mutant exhibited decreased survival after radiation exposure, impaired adhesion to bone marrow stroma cell and decreased bone marrow stroma cell–induced proliferation. UBE2I confers cells with multiple advantages to promote tumorigenesis and predicts decreased survival when combined with PIAS1. The sumoylation pathway is a novel therapeutic target with implications for existing proteasomal-based treatment strategies.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1604-1604
Author(s):  
HoangDinh Huynh ◽  
Junke Zheng ◽  
Chengcheng Zhang

Abstract Abstract 1604 Previously we identified IGFBP2 as an extrinsic factor that supports ex vivo expansion of hematopoietic stem cells (HSCs). The role of IGFBP2 in HSCs and cancer is very intriguing. IGFBP2 can bind to insulin-like growth factor (IGF) ligands and displays IGF-dependent growth inhibitory effects on many cell types. On the other hand, IGFBP2 is capable of stimulating growth of certain cancer cells, and is overexpressed in many cancer patients and its expression is correlated with cancer progression. Here we sought to study the role of IGFBP2 in regulation of the activity of normal HSCs. We showed that IGFBP2 was expressed in differentiated hematopoietic cells and bone marrow stroma but not in HSCs. Consistent with its gene expression pattern, IGFBP2-/- HSCs had similar repopulation activity as their wild-type counterparts. By contrast, when we transplanted HSCs into IGFBP2-/- or wild-type recipient mice, we found decreased in vivo repopulation of HSCs in primary and secondary transplanted IGFBP2-/- recipients, suggesting that the environmental IGFBP2 positively supports HSC activity. Further co-culture of HSCs with IGFBP2-/- or wild-type bone marrow stromal cells indicated that IGFBP2 produced by bone marrow stroma indeed supports HSC expansion. Consistently, HSCs in IGFBP2-/- mice showed decreased frequency and cell cycling, and had upregulated expression of cell cycle inhibitors of p21, p16, and p19. To determine whether IGFBP2's effect on HSCs depends on IGF signaling, we compared the repopulation of donor cells deficient for the IGF type I receptor in wild-type and IGFBP2-/- recipients. These HSCs that are defective in IGF signaling still have decreased repopulation in IGFBP2-/- recipients, suggesting that the environmental effect of IGFBP2 on HSCs is independent of IGF signaling. To identify the functional domain of IGFBP2 in regulation of HSC activity, we constructed IGFBP2 with mutated RGD domain or deleted c-terminus and used the mutant IGFBP2 proteins in ex vivo culture of HSCs. We found that the c-terminus of IGFBP2 is essential to support HSC activity. We are currently in the process of identifying the potential receptor of IGFBP2 on HSCs. In summary, we found that IGFBP2 supports the cycling of normal HSCs, and this effect is independent of IGF signaling. Our study is important in revealing the relationship among environmental cues and cell fates of stem cells and opens up a new avenue in investigation of the roles of IGFBP2 in stem cells and cancer. Disclosures: No relevant conflicts of interest to declare.


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.


2010 ◽  
Vol 207 (7) ◽  
pp. 1359-1367 ◽  
Author(s):  
Ping Lu ◽  
Isaiah L. Hankel ◽  
Judit Knisz ◽  
Andreas Marquardt ◽  
Ming-Yi Chiang ◽  
...  

A recessive mutation named Justy was found that abolishes B lymphopoiesis but does not impair other major aspects of hematopoiesis. Transplantation experiments showed that homozygosity for Justy prevented hematopoietic progenitors from generating B cells but did not affect the ability of bone marrow stroma to support B lymphopoiesis. In bone marrow from mutant mice, common lymphoid progenitors and pre-pro–B cells appeared normal, but cells at subsequent stages of B lymphopoiesis were dramatically reduced in number. Under culture conditions that promoted B lymphopoiesis, mutant pre-pro–B cells remained alive and began expressing the B cell marker CD19 but failed to proliferate. In contrast, these cells were able to generate myeloid or T/NK precursors. Genetic and molecular analysis demonstrated that Justy is a point mutation within the Gon4-like (Gon4l) gene, which encodes a protein with homology to transcriptional regulators. This mutation was found to disrupt Gon4l pre-mRNA splicing and dramatically reduce expression of wild-type Gon4l RNA and protein. Consistent with a role for Gon4l in transcriptional regulation, the levels of RNA encoding C/EBPα and PU.1 were abnormally high in mutant B cell progenitors. Our findings indicate that the Gon4l protein is required for B lymphopoiesis and may function to regulate gene expression during this process.


2008 ◽  
Vol 283 (27) ◽  
pp. 18573-18581 ◽  
Author(s):  
Yasushi Fukaya ◽  
Hiroyuki Shimada ◽  
Ling-Chi Wang ◽  
Ebrahim Zandi ◽  
Yves A. DeClerck

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2378-2378
Author(s):  
Tint Lwin ◽  
Lori A. Hazlehurst ◽  
Lynn C. Moscinski ◽  
William S. Dalton ◽  
Jianguo Tao

Abstract Stromal cells are an essential component of the bone marrow microenvironment that regulate or supports tumor survival. The bone marrow stroma has long been recognized as a "sanctuary site" for lymphoma cells during traditional chemotherapy. In this study, we demonstrated that adhesion of the B-cell lymphoma cell lines SUDH-4 and 10 to bone marrow stroma inhibited mitoxantrone-induced apoptosis. This adhesion-dependent inhibition of mitoxantrone-induced apoptosis correlated with decreased activation of caspases- 8 and 9, and cleavage of caspase 3 and PARP. EMSA analysis demonstrated significantly increased NF-kB binding activity in cells adhered to stroma cells compared to lymphoma cells in suspension. This DNA binding activity could be attributed to cell adhesion-mediated proteolysis of p100 NF-kappaB2 resulted in generation of active p52, which translocates to the nucleus in complex with p65 and RelB. Furthermore, co-culture with stromal cells also induced expression of the NF-kB-regulated anti-apoptotic molecules, XIAP, cIAP1 and cIAP2. In addition, because of the knowledge that BAFF shown to be critical for maintenance of normal B cell homeostasis as well as the survival of malignant B cells, we characterized the functional significance of BAFF in BMS-mediated drug resistance and survival. BAFF was detected on BMS cell line HS-5 and BMS cells derived from lymphoma patients by flow cytometry. Concentrations of BAFF were 3- to 25-fold higher in bone marrow aspirate than in peripheral blood. Lymphoma cell adhesion to HS-5 cells significantly increased BAFF secretion evidenced by both ELISA and immunoblotting. Addition of BAFF counteracted mitoxantrone-induced apoptosis, and elicited a reduction in spontaneous apoptosis in primary lymphomas via activation of NF-kB activation. Finally neutralization of BAFF by TACI enhanced lymphoma cell response to chemotherapy and overcame cell-adhesion mediated drug resistance, suggesting that lymphoma cells utilize BAFF as survival factor. These data indicate that stromal cells prevent apoptosis of lymphoma cells by upregulation of BAFF and induction of NF-kB-regulated anti-apoptotic proteins. Bone marrow-derived BAFF plays a key role in lymphoma cell survival and drug resistance in bone marrow microenvironment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1363-1363
Author(s):  
Junke Zheng ◽  
HoangDinh Huynh ◽  
Chengcheng Zhang

Abstract We previously identified a group of angiopoietin-like proteins (Angptls) as new growth factors that stimulate ex vivo expansion of hematopoietic stem cells (HSCs). To investigate the physiological function of Angptl3 in bone marrow, we characterized the Angptl3 deficient mice, and identified several defects in the hematopoietic compartment. When we transplanted wild-type HSCs into lethally irradiated Angptl3 deficient mice, we found that the mutant bone marrow stroma have much lower ability to support in vivo expansion of HSCs. We sought to identify the Angptl3-producing cells in mouse bone marrow stroma, and showed that Angptl3 is highly expressed in CD45-SSEA4+ cells, which are mesenchymal stem cells (MSCs). Indeed, the co-culture of HSCs with CD45-SSEA4+ MSCs resulted in ex vivo expansion of HSCs. DNA microarray analysis, real-time RT-PCR, and flow cytometry were used to identify the intracellular factors that are responsible for Angptl3’s effects on HSCs. This investigation demonstrated that Angptl3-stimulated HSC expansion is contributed by its activities to support HSC self-renewal and inhibit hematopoietic differentiation. Our study will likely lead to the identification of a novel component of the niche for HSCs.


Sign in / Sign up

Export Citation Format

Share Document