miR-125b and miR-532-3p predict the efficiency of rituximab-mediated lymphodepletion in chronic lymphocytic leukemia patients. A French Innovative Leukemia Organization study

Anne-Laure Gagez, Isabelle Duroux-Richard, Stéphane Leprêtre, Frédérique Orsini-Piocelle, Rémi Letestu, Sophie De Guibert, Edouard Tuaillon, Véronique Leblond, Olfa Khalifa, Valérie Gouilleux-Gruart, Anne Banos, Olivier Tournilhac, Jehan Dupuis, Christian Jorgensen, Guillaume Cartron, Florence Apparailly, Anne-Laure Gagez, Isabelle Duroux-Richard, Stéphane Leprêtre, Frédérique Orsini-Piocelle, Rémi Letestu, Sophie De Guibert, Edouard Tuaillon, Véronique Leblond, Olfa Khalifa, Valérie Gouilleux-Gruart, Anne Banos, Olivier Tournilhac, Jehan Dupuis, Christian Jorgensen, Guillaume Cartron, Florence Apparailly

Abstract

The underlying in vivo mechanisms of rituximab action remain incompletely understood in chronic lymphocytic leukemia. Recent data suggest that circulating micro-ribonucleic acids correlate with chronic lymphocytic leukemia progression and response to rituximab. Our study aimed at identifying circulating micro-ribonucleic acids that predict response to rituximab monotherapy in chronic lymphocytic leukemia patients. Using a hierarchical clustering of micro-ribonucleic acid expression profiles discriminating 10 untreated patients with low or high lymphocyte counts, we found 26 micro-ribonucleic acids significantly deregulated. Using individual real-time reverse transcription polymerase chain reaction, the expression levels of micro-ribonucleic acids representative of these two clusters were further validated in a larger cohort (n=61). MiR-125b and miR-532-3p were inversely correlated with rituximab-induced lymphodepletion (P=0.020 and P=0.001, respectively) and with the CD20 expression on CD19+ cells (P=0.0007 and P<0.0001, respectively). In silico analyses of genes putatively targeted by both micro-ribonucleic acids revealed a central role of the interleukin-10 pathway and CD20 (MS4A1) family members. Interestingly, both micro-ribonucleic acids were negatively correlated with MS4A1 expression, while they were positively correlated with MS4A3 and MSA47 Our results identify novel circulating predictive biomarkers for rituximab-mediated lymphodepletion efficacy in chronic lymphocytic leukemia, and suggest a novel molecular mechanism responsible for the rituximab mode of action that bridges miR-125b and miR-532-3p and CD20 family members. (clinicaltrials.gov Identifier: 01370772).

Trial registration: ClinicalTrials.gov NCT01370772.

Copyright© Ferrata Storti Foundation.

Figures

Figure 1.
Figure 1.
MicroRNAs expression profile discriminates CLL patients with low or high lymphocyte counts before treatment. (A) The profiles of 26 microRNAs significantly differently expressed (P<0.05) between high and low lymphocyte concentration samples isolated from CLL patients (n=5/group) were visualized using a supervised heatmap (average linkage and Pearson’s correlation). Relative miRNA expression was calculated using the comparative threshold cycle (Ct) method. For normalization, the mean Ct value of all miRNA targets was used. Dendrograms indicated the correlation between groups of samples and miRNAs. Samples are in columns and miRNAs in rows. Each column represents an individual sample and each row represents a single miRNA. The heat map shows relative levels of miRNA expression in a green (low relative expression) to red (high relative expression) scale. (B–C) Expression levels of 4 miRNAs representative of each cluster, miR-193 band miR-125b for cluster 1 (B), and miR-532-3p and miR-652 for cluster 2 (C), were measured for 123 CLL patients included in the CLL2010FMP protocol, using RT-qPCR. A significant inverse correlation was observed depending on the lymphocyte counts for miR-193b r=−0.19), miR-125b (r=−0.39), miR-652 (r=−0.30) and miR-532-3p (r=−0.34) (Spearman’s correlation test).
Figure 2.
Figure 2.
Efficacy of lymphodepletion after rituximab treatment inversely correlates with miR-125b and miR-532-3p expression levels. miR-125b (r=−0.42) (A) and miR-532-3p (r=−0.49) (B) expression levels were quantified by RT-qPCR. The lymphodepletion was measured 22 days after 4 doses of rituximab infusion and correlated with miRNAs (Spearman’s correlation test) (n=61).
Figure 3.
Figure 3.
Target gene prediction for miR-125b and miR-532-3p. (A) To identify putative miR-125b and miR-532-3p target genes, we used miRWalk software. VENNY, an interactive tool for comparing lists with Venn Diagrams, was used to predict common genes between human regulatory B cells IL-10+ and IL-10−, and putative miR-125b and miR-532-3p target genes. 26 genes putatively targeted by miR-125b and miR-532-3p, and specifically dysregulated in IL-10+ B cells transcriptome analysis are listed. (B) The layout of these 26 putative targets was built in the context of rituximab treatment (MS4A1 (CD20 gene)) using Ingenuity Pathway Analysis (IPA). 9 genes revealed a central role for the IL-10 pathway.
Figure 4.
Figure 4.
Inverse correlation between miR-125b and miR-532-3p expression levels and circulating CD20 surface antigen expression on CD19+ cells in CLL patients blood. CD20 expression levels on CD19+ lymphocytes were quantified using flow cytometry. PBMCs were collected and miR-125b (r=−0.37) (A) and miR-532-3p (r=−0.29) (B) were quantified using RT-qPCR. Circulating CD20 antigen was correlated to miRNA expression (Spearman’s correlation test) (n=61).
Figure 5.
Figure 5.
miR-125b and miR-532-3p expression levels correlate with the expression of three MS4A family members. (A) Spearman’s correlation between miR-125b or miR-532-3p and MS4A mRNA family expression levels in CLL patients blood (n=61). (B) Putative miR-125b and miR-532-3p target binding sites on MS4A family members. CDS: coding DNA sequence; rho: correlation coefficient; UTR: untranslated region.
Figure 6.
Figure 6.
Schematic representation of the potential mechanisms of action of miR-125b and miR-532-3p on the modulation of rituximab activity in CLL cells. In CLL patients displaying low expression levels of miR-125b and miR-532-3p, the MS4A1 mRNA and CD20 surface receptor are upregulated, while the expression of MS4A3 and MS4A7 mRNA are downregulated (MS4A3 and MS4A7 protein expressions were unknown). Consequently, rituximab (RTX) efficacy is optimum for lymphodepletion (Lymphodepletion +++). In contrast, CLL patients with high expression levels of miR-125b and miR-532-3p display low levels of MS4A1 mRNA and CD20 surface receptor, and high levels of MS4A3 and MS4A7 mRNA (MS4A3 and MS4A7 protein expressions were unknown), which might form hetero-oligomers with CD20 and impede optimal lymphodepletion (Lymphodepletion +) by rituximab. Interrogation marks indicate data that were not experimentally confirmed in the present study.

References

    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–233.
    1. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006; 6(11):857–866.
    1. Lawrie CH. MicroRNAs in hematological malignancies. Blood rev. 2013;27(3):143–154.
    1. Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009;10(10):704–714.
    1. Li PP, Wang X. Role of signaling pathways and miRNAs in chronic lymphocytic leukemia. Chin Med J (Engl). 2013; 126(21): 4175–4182.
    1. Tavolaro S, Colombo T, Chiaretti S, et al. Increased chronic lymphocytic leukemia proliferation upon IgM stimulation is sustained by the upregulation of miR-132 and miR-212. Genes Chromosomes Cancer. 2015;54(4):222–234.
    1. Calin GA, Dumitru CD, Shimizu M, et al. Frequent deletions and downregulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2002; 99(24):15524–15529.
    1. Palacios F, Abreu C, Prieto D, et al. Activation of the PI3K/AKT pathway by microRNA-22 results in CLL B-cell proliferation. Leukemia. 2015;29(1):115–125.
    1. Rushworth SA, Murray MY, Barrera LN, Heasman SA, Zaitseva L, Macewan DJ. Understanding the role of miRNA in regulating NF-kappaB in blood cancer. Am J Cancer Res. 2012;2(1):65–74.
    1. Bomben R, Gobessi S, Dal Bo M, et al. The miR-17 approximately 92 family regulates the response to Toll-like receptor 9 triggering of CLL cells with unmutated IGHV genes. Leukemia. 2012;26(7):1584–1593.
    1. Cui B, Chen L, Zhang S, et al. MicroRNA-155 influences B-cell receptor signaling and associates with aggressive disease in chronic lymphocytic leukemia. Blood. 2014; 124(4):546–554.
    1. Mraz M, Chen L, Rassenti LZ, et al. miR-150 influences B-cell receptor signaling in chronic lymphocytic leukemia by regulating expression of GAB1 and FOXP1. Blood. 2014;124(1):84–95.
    1. Cimmino A, Calin GA, Fabbri M, et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci USA. 2005;102(39):13944–13949.
    1. Pekarsky Y, Santanam U, Cimmino A, et al. Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res. 2006;66(24):11590–11593.
    1. Mraz M, Malinova K, Kotaskova J, et al. miR-34a, miR-29c and miR-17-5p are downregulated in CLL patients with TP53 abnormalities. Leukemia. 2009;23(6):1159–1163.
    1. Fabbri M, Bottoni A, Shimizu M, et al. Association of a microRNA/TP53 feedback circuitry with pathogenesis and outcome of B-cell chronic lymphocytic leukemia. JAMA. 2011;305(1):59–67.
    1. Zou ZJ, Fan L, Wang L, et al. miR-26a and miR-214 down-regulate expression of the PTEN gene in chronic lymphocytic leukemia, but not PTEN mutation or promoter methylation. Oncotarget. 2015; 6(2):1276–1285.
    1. Ferracin M, Zagatti B, Rizzotto L, et al. MicroRNAs involvement in fludarabine refractory chronic lymphocytic leukemia. Mol Cancer. 2010;9:123.
    1. Moussay E, Palissot V, Vallar L, et al. Determination of genes and microRNAs involved in the resistance to fludarabine in vivo in chronic lymphocytic leukemia. Mol Cancer. 2010;9:115.
    1. Zenz T, Habe S, Denzel T, et al. Detailed analysis of p53 pathway defects in fludarabine-refractory chronic lymphocytic leukemia (CLL): dissecting the contribution of 17p deletion, TP53 mutation, p53-p21 dysfunction, and miR34a in a prospective clinical trial. Blood. 2009;114(13):2589–2597.
    1. De Tullio G, De Fazio V, Sgherza N, et al. Challenges and opportunities of microRNAs in lymphomas. Molecules. 2014;19(9):14723–14781.
    1. Gilad S, Meiri E, Yogev Y, et al. Serum microRNAs are promising novel biomarkers. PloS One. 2008;3(9):e3148.
    1. Mitchell PS, Parkin RK, Kroh EM, et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105(30):10513–10518.
    1. Visone R, Veronese A, Rassenti LZ, et al. miR-181b is a biomarker of disease progression in chronic lymphocytic leukemia. Blood. 2011;118(11):3072–3079.
    1. Duroux-Richard I, Pers YM, Fabre S, et al. Circulating miRNA-125b is a potential biomarker predicting response to rituximab in rheumatoid arthritis. Mediators Inflamm. 2014;2014:342524.
    1. Rossi RL, Rossetti G, Wenandy L, et al. Distinct microRNA signatures in human lymphocyte subsets and enforcement of the naive state in CD4+ T cells by the microRNA miR-125b. Nat Immunol. 2011; 12(8):796–803.
    1. Ooi AG, Sahoo D, Adorno M, Wang Y, Weissman IL, Park CY. MicroRNA-125b expands hematopoietic stem cells and enriches for the lymphoid-balanced and lymphoid-biased subsets. Proc Natl Acad Sci USA. 2010;107(50):21505–21510.
    1. Tili E, Michaille JJ, Luo Z, et al. The down-regulation of miR-125b in chronic lymphocytic leukemias leads to metabolic adaptation of cells to a transformed state. Blood. 2012;120(13):2631–2638.
    1. Cartron G, Trappe RU, Solal-Celigny P, Hallek M. Interindividual variability of response to rituximab: from biological origins to individualized therapies. Clin Cancer Res. 2011;17(1):19–30.
    1. Lepretre S, Letestu R, Dartigeas C, et al. Results of a Phase II Randomizing Intensified Rituximab Pre-Phase Followed By Standard FCR Vs Standard FCR in Previously Untreated Patients with Active B-Chronic Lymphocytic Leukemia (B-CLL). CLL2010FMP (for fit medically patients): A Study of the french Cooperative Group on CLL and WM (FCGCLL/MW) and the “Groupe Ouest-Est d’Etudes Des Leucémies Aigües Et Autres Maladies Du sang” (GOELAMS). Blood. 2014;124(21): 3329.
    1. Li J, Zhi J, Wenger M, et al. Population pharmacokinetics of rituximab in patients with chronic lymphocytic leukemia. J Clin Pharmacol. 2012;52(12):1918–1926.
    1. Hallek M, Cheson BD, Catovsky D, et al. Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. Blood. 2008; 111(12):5446–5456.
    1. Iwata Y, Matsushita T, Horikawa M, et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood. 2011; 117(2):530–541.
    1. Gagez AL, Tuaillon E, Cezar R, et al. Response to rituximab in B-CLL patients is adversely impacted by frequency of IL-10 competent B cells and FcgammaRIIIa polymorphism. A study of FCGCLL/WM and GOELAMS groups. Blood Cancer J. 2016; 6:e389.
    1. Negrini M, Cutrona G, Bassi C, et al. microRNAome expression in chronic lymphocytic leukemia: comparison with normal B-cell subsets and correlations with prognostic and clinical parameters. Clin Cancer Res. 2014;20(15):4141–4153.
    1. Moussay E, Wang K, Cho JH, et al. MicroRNA as biomarkers and regulators in B-cell chronic lymphocytic leukemia. Proc Natl Acad Sci USA. 2011;108(16):6573–6578.
    1. Dweep H, Gretz N. miRWalk2.0: a comprehensive atlas of microRNA-target interactions. Nat Methods. 2015;12(8):697.
    1. Oliveros JC. (2007) VENNY. An interactive tool for comparing lists with Venn Diagrams. .
    1. Lin W, Cerny D, Chua E, et al. Human regulatory B cells combine phenotypic and genetic hallmarks with a distinct differentiation fate. J Immunol. 2014;193(5):2258–2266.
    1. So AY, Sookram R, Chaudhuri AA, et al. Dual mechanisms by which miR-125b represses IRF4 to induce myeloid and B-cell leukemias. Blood. 2014;124(9):1502–1512.
    1. Sonoki T, Iwanaga E, Mitsuya H, Asou N. Insertion of microRNA-125b-1, a human homologue of lin-4, into a rearranged immunoglobulin heavy chain gene locus in a patient with precursor B-cell acute lymphoblastic leukemia. Leukemia. 2005; 19(11):2009–2010.
    1. Malumbres R, Sarosiek KA, Cubedo E, et al. Differentiation stage-specific expression of microRNAs in B lymphocytes and diffuse large B-cell lymphomas. Blood. 2009; 113(16):3754–3764.
    1. Gururajan M, Haga CL, Das S, et al. MicroRNA 125b inhibition of B cell differentiation in germinal centers. Int Immunol. 2010;22(7):583–592.
    1. Ruiz-Lafuente N, Alcaraz-Garcia MJ, Sebastian-Ruiz S, et al. The gene expression response of chronic lymphocytic leukemia cells to IL-4 is specific, depends on ZAP-70 status and is differentially affected by an NFkappaB inhibitor. PloS One. 2014; 9(10):e109533.
    1. Ruiz-Lafuente N, Alcaraz-Garcia MJ, Sebastian-Ruiz S, et al. IL-4 up-regulates MiR-21 and the MiRNAs hosted in the CLCN5 gene in chronic lymphocytic leukemia. PloS One. 2015;10(4):e0124936.
    1. Clark EA, Ledbetter JA. How does B cell depletion therapy work, and how can it be improved? Ann Rheum Dis. 2005;64 Suppl 4:iv77–80.
    1. Liang Y, Tedder TF. Identification of a CD20-, FcepsilonRIbeta-, and HTm4-related gene family: sixteen new MS4A family members expressed in human and mouse. Genomics. 2001;72(2):119–127.
    1. Kutok JL, Yang X, Folkerth R, Adra CN. Characterization of the expression of HTm4 (MS4A3), a cell cycle regulator, in human peripheral blood cells and normal and malignant tissues. J Cell Mol Med. 2011;15(1):86–93.
    1. Gingras MC, Lapillonne H, Margolin JF. CFFM4: a new member of the CD20/FcepsilonRIbeta family. Immunogenetics. 2001;53(6):468–476.
    1. Banzhaf-Strathmann J, Edbauer D. Good guy or bad guy: the opposing roles of microRNA 125b in cancer. Cell Commun Signal. 2014;12:30.

Source: PubMed

3
Abonneren