miRNA profile at diagnosis predicts treatment outcome in patients with B-chronic lymphocytic leukemia: A FILO study

Isabelle Duroux-Richard, Anne-Laure Gagez, Elina Alaterre, Rémi Letestu, Olfa Khalifa, Christian Jorgensen, Stéphane Leprêtre, Emmanuelle Tchernonog, Jérôme Moreaux, Guillaume Cartron, Florence Apparailly, Isabelle Duroux-Richard, Anne-Laure Gagez, Elina Alaterre, Rémi Letestu, Olfa Khalifa, Christian Jorgensen, Stéphane Leprêtre, Emmanuelle Tchernonog, Jérôme Moreaux, Guillaume Cartron, Florence Apparailly

Abstract

During many years, chemo-immunotherapy fludarabine-cyclophosphamide-rituximab (FCR) was the gold standard for first line treatment of medically fit patients with symptomatic B-chronic lymphocytic leukemia (CLL). Over the last decade, targeted biotherapies have revolutionized the treatment of B-CLL patients and almost entirely supplanted FCR. However, no biomarker still exists to predict the complete remission (CR) with undetectable minimal residual disease (uMRD) in bone marrow (BM), which remains the best predictive factor for survival. MicroRNAs represent a class of molecular biomarkers which expression is altered in B-CLL. Our study aimed at identifying before treatment blood miRNAs that predict treatment outcome in previously untreated B-CLL patients (NCT01370772, https://ichgcp.net/clinical-trials-registry/NCT01370772). Using hierarchical clustering of miRNA expression profiles discriminating 8 patients who achieved CR with BM uMRD from 8 patients who did not achieve CR and displayed detectable BM MRD, we identified 25 miRNAs differentially expressed before treatment. The expression of 11 miRNAs was further validated on a larger cohort (n=123). Based on the dosage of 5 miRNAs at diagnosis, a decision tree was constructed to predict treatment outcome. We identified 6 groups of patients with a distinct probability of being CR with BM uMRD to FCR treatment, ranging from 72% (miR-125b, miR-15b and miR-181c high) to 4% (miR-125b and miR-193b low). None of the patients displaying high expression levels of miR-125b, miR-15b and miR-181c relapsed during study follow-up. In contrast, patients with low miR-15b and high miR-412, or with low miR-125b and miR-193b, demonstrated significant low PFS. RNA sequencing of blood at diagnosis identified that patients relapsing after treatment are characterized by significant enrichment of gene signatures related to cell cycle, MYC target genes, metabolism and translation regulation. Conversely, patients achieving CR with BM uMRD displayed significant enrichment in genes related to communication between CLL cells and the microenvironment, immune system activation and upregulation of polycomb PRC2 complex target genes. Our results suggest that blood miRNAs are potent predictive biomarkers for FCR treatment efficacy and might be implicated in the FCR efficacy in B-CLL patients, providing new insight into unmet need for the treatment of B-CLL patients and identifying pathways predictive of patients' remission.

Clinical trial registration: ClinicalTrials.gov, identifier NCT01370772.

Keywords: B-CLL; diagnosis; microRNA; prediction; trancriptomics; treatment outcome.

Conflict of interest statement

Author GC received consultancy by Roche and BMS and honoraria from Roche, BMS, Jansen, Novartis and Gilead. Author ET received consultancy and honoraria from Janssen and Abbvie. Author SL received honoraria from Gilead, Janssen, Beigene, Abbvie, Astra Zeenca. Author OK was employed by Erasmus Mundus (2013-2016). The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be constructed as a potential conflict of interest.

Copyright © 2022 Duroux-Richard, Gagez, Alaterre, Letestu, Khalifa, Jorgensen, Leprêtre, Tchernonog, Moreaux, Cartron and Apparailly.

Figures

Figure 1
Figure 1
miRNAs associated with complete remission and uMRD (A) miRNAs associated with CR and BM uMRD (undetectable minimal residual disease) in previously untreated B-CLL patients. A volcano plot compares the normalized expression of miRNAs in first line of treatment outcome in CR with blood and BM uMRD patients (n=8) versus no-CR with detectable MRD value patients (n=8) assessed three months after treatment. Log2 fold changes and their corresponding P-values of all genes in the microarray were taken for construction of the volcano plot. The y-axis is the negative log10 of P values (a higher value indicates greater significance) and the x-axis is the difference in expression between two experimental groups. Genes up-regulated with more than 2.5-fold change are depicted in red boxes and those downregulated with identical fold change are in green boxes. Genes above the blue line are significantly deregulated (P-value<0.05, t-test). All other genes in the array that were not deregulated are in black dots. 25 genes were identified as significantly up or downregulated. (B) The expression level of 11 miRNAs was quantified using multiplex RT-qPCR in a validation cohort of treatment-naïve B-CLL patients (n=100). R: responders, NR: non-responders. Median are plotted, * P-value<0.05.
Figure 2
Figure 2
A decision-making model using expression of 5 blood miRNAs at baseline to classify B-CLL patients and predict treatment outcome. Receiver Operating Characteristic (ROC) curves method was used to constructed the decision tree, underlying 6 groups of patients. For each group, relative expression of 2 or 3 miRNAs into the blood of B-CLL patients (n=100) can give the probability at diagnosis of being in CR with BM uMRD three months treatment termination. Red and green colors were used for high (H) and low (L) miRNA expression levels.
Figure 3
Figure 3
Progression free survival (PFS) in the total cohort according to the five-miRNA decision tree signature. PFS in the total cohort (A), according to the six different groups of signatures identified in the miRNA decision tree (B), or with combined miRNA signature groups (C). PFS, Progression free survival; HHH, group of patients with miR-125b high, miR-15b high and miR-181c high; HHL, group of patients with miR-125b high, miR-15b high and miR-181c low; HLH, group of patients with miR-125b high, miR-15b low and miR-412 high; HLL, group of patients with miR-125b high, miR-15b low and miR-412 low; LH, group of patients with miR-125b low and miR-193b high; LL, group of patients with miR-125b low and miR-193b low; HHL+HLH+HLL+LH, consolidation of HHL-, HLH-, HLL-, and LH-group.
Figure 4
Figure 4
Gene expression profiling of three prognostic groups defined by the five-miRNA decision tree signature. Clustergram of the 1373 differentially expressed genes between HHH group and HLH and LL groups (FDR (A). Molecular signatures enriched in HHH group (B) or HLH and LL groups (C) using GSEA Database (all curated gene sets), and relevant pathways are presented (FDR q value ≤ 0.05).

References

    1. Rozman C, Montserrat E. Chronic lymphocytic leukemia. N Engl J Med (1995) 333(16):1052−7. doi: 10.1056/NEJM199510193331606
    1. Robak T, Dmoszynska A, Solal-Céligny P, Warzocha K, Loscertales J, Catalano J, et al. . Rituximab plus fludarabine and cyclophosphamide prolongs progression-free survival compared with fludarabine and cyclophosphamide alone in previously treated chronic lymphocytic leukemia. JCO (2010) 28(10):1756−65. doi: 10.1200/JCO.2009.26.4556
    1. Goede V, Fischer K, Engelke A, Schlag R, Lepretre S, Montero LFC, et al. . Obinutuzumab as frontline treatment of chronic lymphocytic leukemia: updated results of the CLL11 study. Leukemia (2015) 29(7):1602−4.
    1. Goede V, Fischer K, Busch R, Jaeger U, Dilhuydy MS, Wickham N, et al. . Chemoimmunotherapy with GA101 plus chlorambucil in patients with chronic lymphocytic leukemia and comorbidity: results of the CLL11 (BO21004) safety run-in. Leukemia (2013) 27(5):1172−4.
    1. Brown JR. Ibrutinib (PCI-32765), the first BTK (Bruton’s tyrosine kinase) inhibitor in clinical trials. Curr Hematol Malig Rep (2013) 8(1):1−6. doi: 10.1007/s11899-012-0147-9
    1. Thompson PA, Stingo F, Keating MJ, Ferrajoli A, Burger JA, Wierda WG, et al. . Outcomes of patients with chronic lymphocytic leukemia treated with first-line idelalisib plus rituximab after cessation of treatment for toxicity: Outcome after ending idelalisib in CLL. Cancer (2016) 122(16):2505−11. doi: 10.1002/cncr.30069
    1. Itchaki G, Brown JR. Experience with ibrutinib for first-line use in patients with chronic lymphocytic leukemia. Ther Adv Hematol (2018) 9(1):3−19. doi: 10.1177/2040620717741861
    1. Baliakas P, Mattsson M, Stamatopoulos K, Rosenquist R. Prognostic indices in chronic lymphocytic leukaemia: where do we stand how do we proceed? J Intern Med (2016) 279(4):347−57. doi: 10.1111/joim.12455
    1. Delgado J, Salaverria I, Baumann T, Martinez-Trillos A, Lee E, Jimenez L, et al. . Genomic complexity and IGHV mutational status are key predictors of outcome of chronic lymphocytic leukemia patients with TP53 disruption. Haematologica (2014) 99(11):e231−4. doi: 10.3324/haematol.2014.108365
    1. Farren TW, Giustiniani J, Fanous M, Liu F, Macey MG, Wright F, et al. . Minimal residual disease detection with tumor-specific CD160 correlates with event-free survival in chronic lymphocytic leukemia. Blood Cancer J (2015) 5(1):e273−e273.
    1. Böttcher S, Ritgen M, Fischer K, Stilgenbauer S, Busch RM, Fingerle-Rowson G, et al. . Minimal residual disease quantification is an independent predictor of progression-free and overall survival in chronic lymphocytic leukemia: A multivariate analysis from the randomized GCLLSG CLL8 trial. JCO (2012) 30(9):980−8. doi: 10.1200/JCO.2011.36.9348
    1. Letestu R, Dahmani A, Boubaya M, Baseggio L, Campos L, Chatelain B, et al. . Prognostic value of high-sensitivity measurable residual disease assessment after front-line chemoimmunotherapy in chronic lymphocytic leukemia. Leukemia (2021) 35(6):1597−609.
    1. Thompson PA, Wierda WG. Eliminating minimal residual disease as a therapeutic end point: working toward cure for patients with CLL. Blood (2016) 127(3):279−86.
    1. Thompson PA, Tam CS, O’Brien SM, Wierda WG, Stingo F, Plunkett W, et al. . Fludarabine, cyclophosphamide, and rituximab treatment achieves long-term disease-free survival in IGHV-mutated chronic lymphocytic leukemia. Blood (2016) 127(3):303−9.
    1. Van Roosbroeck K, Calin GA. MicroRNAs in chronic lymphocytic leukemia: miRacle or miRage for prognosis and targeted therapies? Semin Oncol (2016) 43(2):209−14.
    1. Ferracin M, Zagatti B, Rizzotto L, Cavazzini F, Veronese A, Ciccone M, et al. . MicroRNAs involvement in fludarabine refractory chronic lymphocytic leukemia. Mol Cancer (2010) 9(1):123. doi: 10.1186/1476-4598-9-123
    1. Liang H, Gong F, Zhang S, Zhang CY, Zen K, Chen X. The origin, function, and diagnostic potential of extracellular microRNAs in human body fluids: Extracellular microRNAs in human body fluids. WIREs RNA (2014) 5(2):285−300. doi: 10.1002/wrna.1208
    1. De Tullio G, De Fazio V, Sgherza N, Minoia C, Serratì S, Merchionne F, et al. . Challenges and opportunities of MicroRNAs in lymphomas. Molecules (2014) 19(9):14723−81.
    1. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, et al. . Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA (2008) 105(30):10513−8. doi: 10.1073/pnas.0804549105
    1. Moussay E, Wang K, Cho JH, van Moer K, Pierson S, Paggetti J, et al. . MicroRNA as biomarkers and regulators in b-cell chronic lymphocytic leukemia. Proc Natl Acad Sci USA (2011) 108(16):6573−8. doi: 10.1073/pnas.1019557108
    1. Ferrajoli A, Shanafelt TD, Ivan C, Shimizu M, Rabe KG, Nouraee N, et al. . Prognostic value of miR-155 in individuals with monoclonal b-cell lymphocytosis and patients with b chronic lymphocytic leukemia. Blood (2013) 122(11):1891−9.
    1. Gagez AL, Duroux-Richard I, Leprêtre S, Orsini-Piocelle F, Letestu R, De Guibert S, et al. . miR-125b and miR-532-3p predict the efficiency of rituximab-mediated lymphodepletion in chronic lymphocytic leukemia patients. a French innovative leukemia organization study. Haematologica (2017) 102(4):746−54. doi: 10.3324/haematol.2016.153189
    1. Visone R, Veronese A, Rassenti LZ, Balatti V, Pearl DK, Acunzo M, et al. . miR-181b is a biomarker of disease progression in chronic lymphocytic leukemia. Blood (2011) 118(11):3072−9.
    1. Hallek M, Cheson BD, Catovsky D, Caligaris-Cappio F, Dighiero G, Döhner H, 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):5446−56.
    1. Cartron G, Letestu R, Dartigeas C, Tout M, Mahé B, Gagez AL, et al. . Increased rituximab exposure does not improve response and outcome of patients with chronic lymphocytic leukemia after fludarabine, cyclophosphamide, rituximab. a French innovative leukemia organization (FILO) study. Haematologica (2018) 103(8):e356−9. doi: 10.3324/haematol.2017.182352
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics (2013) 29(1):15−21.
    1. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. . Bioconductor: open software development for computational biology and bioinformatics. Genome Biol (2004) 5(10):R80. doi: 10.1186/gb-2004-5-10-r80
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol (2014) 15(12):550. doi: 10.1186/s13059-014-0550-8
    1. Youden WJ. Index for rating diagnostic tests. Cancer (1950) 3(1):32−5.
    1. Deng Y, Li X, Feng J, Zhang X. Overexpression of miR-202 resensitizes imatinib resistant chronic myeloid leukemia cells through targetting hexokinase 2. Biosci Rep (2018) 38(3):BSR20171383.
    1. Kaur G, Ruhela V, Rani L, Gupta A, Sriram K, Gogia A, et al. . RNA-Seq profiling of deregulated miRs in CLL and their impact on clinical outcome. Blood Cancer J (2020) 10(1):6.
    1. Negrini M, Cutrona G, Bassi C, Fabris S, Zagatti B, Colombo M, 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):4141−53.
    1. Papageorgiou SG, Diamantopoulos MA, Kontos CK, Bouchla A, Vasilatou D, Bazani E, et al. . MicroRNA-92a-3p overexpression in peripheral blood mononuclear cells is an independent predictor of prolonged overall survival of patients with chronic lymphocytic leukemia. Leukemia Lymphoma (2019) 60(3):658−67. doi: 10.1080/10428194.2018.1461861
    1. Bomben R, Gobessi S, Dal Bo M, Volinia S, Marconi D, Tissino E, et al. . The miR-17∼92 family regulates the response to toll-like receptor 9 triggering of CLL cells with unmutated IGHV genes. Leukemia (2012) 26(7):1584−93.
    1. Dal Bo M, Bomben R, Hernández L, Gattei V, The MYC. miR-17-92 axis in lymphoproliferative disorders: A common pathway with therapeutic potential. Oncotarget (2015) 6(23):19381−92. doi: 10.18632/oncotarget.4574
    1. Olive V, Sabio E, Bennett, Margaux J, De Jong, Caitlin S, et al. . A component of the mir-17-92 polycistronic oncomir promotes oncogene-dependent apoptosis. eLife (2013) 2:e00822.
    1. Sbarrato T, Horvilleur E, Pöyry T, Hill K, Chaplin LC, Spriggs RV, et al. . A ribosome-related signature in peripheral blood CLL b cells is linked to reduced survival following treatment. Cell Death Dis (2016) 7(6):e2249−e2249.
    1. Kosalai ST, Morsy MHA, Papakonstantinou N, Mansouri L, Stavroyianni N, Kanduri C, et al. . EZH2 upregulates the PI3K/AKT pathway through IGF1R and MYC in clinically aggressive chronic lymphocytic leukaemia. Epigenetics (2019) 14(11):1125−40.
    1. Alzrigat M, Jernberg-Wiklund H. The miR-125a and miR-320c are potential tumor suppressor microRNAs epigenetically silenced by the polycomb repressive complex 2 in multiple myeloma. RNA Dis (2017) 4(2):e1529.
    1. Svanberg R, Janum S, Patten PEM, Ramsay AG, Niemann CU. Targeting the tumor microenvironment in chronic lymphocytic leukemia. haematol (2021) 106(9):2312−24.
    1. Kassambara A, Jourdan M, Bruyer A, Robert N, Pantesco V, Elemento O, et al. . Global miRNA expression analysis identifies novel key regulators of plasma cell differentiation and malignant plasma cell. Nucleic Acids Res (2017) 45(10):5639−52. doi: 10.1093/nar/gkx327
    1. Zhou X, Franklin RA, Adler M, Jacox JB, Bailis W, Shyer JA, et al. . Circuit design features of a stable two-cell system. Cell (2018) 172(4):744–757.e17.
    1. Allegra D, Bilan V, Garding A, Döhner H, Stilgenbauer S, Kuchenbauer F, et al. . Defective DROSHA processing contributes to downregulation of MiR-15/-16 in chronic lymphocytic leukemia. Leukemia (2014) 28(1):98−107.
    1. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, et al. . Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci USA (2002) 99(24):15524−9. doi: 10.1073/pnas.242606799
    1. Lovat F, Fassan M, Gasparini P, Rizzotto L, Cascione L, Pizzi M, et al. . miR-15b/16-2 deletion promotes b-cell malignancies. Proc Natl Acad Sci USA (2015) 112(37):11636−41. doi: 10.1073/pnas.1514954112
    1. Zanette DL, Rivadavia F, Molfetta GA, Barbuzano FG, Proto-Siqueira R, Falcão RP, et al. . miRNA expression profiles in chronic lymphocytic and acute lymphocytic leukemia. Braz J Med Biol Res (2007) 40(11):1435−40.
    1. Tili E, Michaille JJ, Luo Z, Volinia S, Rassenti LZ, Kipps TJ, 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):2631−8.
    1. Pekarsky Y, Santanam U, Cimmino A, Palamarchuk A, Efanov A, Maximov V, et al. . Tcl1 expression in chronic lymphocytic leukemia is regulated by miR-29 and miR-181. Cancer Res (2006) 66(24):11590−3.
    1. Chaudhuri AA, So AYL, Mehta A, Minisandram A, Sinha N, Jonsson VD, et al. . Oncomir miR-125b regulates hematopoiesis by targeting the gene Lin28A. Proc Natl Acad Sci USA (2012) 109(11):4233−8. doi: 10.1073/pnas.1200677109
    1. Bousquet M, Harris MH, Zhou B, Lodish HF. MicroRNA miR-125b causes leukemia. Proc Natl Acad Sci USA (2010) 107(50):21558−63. doi: 10.1073/pnas.1016611107
    1. Gao Xn, Lin J, Gao L, Li Yh, Wang L L, Yu L. MicroRNA-193b regulates c-kit proto-oncogene and represses cell proliferation in acute myeloid leukemia. Leukemia Res (2011) 35(9):1226−32.
    1. González-Gugel E, Villa-Morales M, Santos J, Bueno MJ, Malumbres M, Rodríguez-Pinilla SM, et al. . Down-regulation of specific miRNAs enhances the expression of the gene smoothened and contributes to T-cell lymphoblastic lymphoma development. Carcinogenesis (2013) 34(4):902−8. doi: 10.1093/carcin/bgs404
    1. Mets E, van der Meulen J, Van Peer G, Boice M, Mestdagh P, Van de Walle I, et al. . MicroRNA-193b-3p acts as a tumor suppressor by targeting the MYB oncogene in T-cell acute lymphoblastic leukemia. Leukemia (2015) 29(4):798−806.
    1. Arribas AJ, Gómez-Abad C, Sánchez-Beato M, Martinez N, DiLisio L, Casado F, et al. . Splenic marginal zone lymphoma: comprehensive analysis of gene expression and miRNA profiling. Mod Pathol (2013) 26(7):889−901.
    1. Kminkova J, Mraz M, Zaprazna K, Navrkalova V, Tichy B, Plevova K, et al. . Identification of novel sequence variations in microRNAs in chronic lymphocytic leukemia. Carcinogenesis (2014) 35(5):992−1002. doi: 10.1093/carcin/bgt396

Source: PubMed

3
订阅