Integrative prognostic models predict long-term survival after immunochemotherapy in chronic lymphocytic leukemia patients

Johannes Bloehdorn, Julia Krzykalla, Karlheinz Holzmann, Andreas Gerhardinger, Billy Michael Chelliah Jebaraj, Jasmin Bahlo, Kathryn Humphrey, Eugen Tausch, Sandra Robrecht, Daniel Mertens, Christof Schneider, Kirsten Fischer, Michael Hallek, Hartmut Döhner, Axel Benner, Stephan Stilgenbauer, Johannes Bloehdorn, Julia Krzykalla, Karlheinz Holzmann, Andreas Gerhardinger, Billy Michael Chelliah Jebaraj, Jasmin Bahlo, Kathryn Humphrey, Eugen Tausch, Sandra Robrecht, Daniel Mertens, Christof Schneider, Kirsten Fischer, Michael Hallek, Hartmut Döhner, Axel Benner, Stephan Stilgenbauer

Abstract

Chemoimmunotherapy with fludarabine, cyclophosphamide and rituximab (FCR) can induce long-term remissions in patients with chronic lymphocytic leukemia. Treatment efficacy with Bruton's tyrosine kinase inhibitors was found similar to FCR in untreated chronic lymphocytic leukemia patients with a mutated immunoglobulin heavy chain variable (IGHV) gene. In order to identify patients who specifically benefit from FCR, we developed integrative models including established prognostic parameters and gene expression profiling (GEP). GEP was conducted on n=337 CLL8 trial samples, "core" probe sets were summarized on gene levels and RMA normalized. Prognostic models were built using penalized Cox proportional hazards models with the smoothly clipped absolute deviation penalty. We identified a prognostic signature of less than a dozen genes, which substituted for established prognostic factors, including TP53 and IGHV gene mutation status. Independent prognostic impact was confirmed for treatment, β2-microglobulin and del(17p) regarding overall survival and for treatment, del(11q), del(17p) and SF3B1 mutation for progression-free survival. The combination of independent prognostic and GEP variables performed equal to models including only established non-GEP variables. GEP variables showed higher prognostic accuracy for patients with long progression-free survival compared to categorical variables like the IGHV gene mutation status and reliably predicted overall survival in CLL8 and an independent cohort. GEP-based prognostic models can help to identify patients who specifically benefit from FCR treatment. The CLL8 trial is registered under EUDRACT-2004- 004938-14 and clinicaltrials gov. Identifier: NCT00281918.

Figures

Figure 1.
Figure 1.
Prediction error estimates for prognostic model combinations. Prediction error curves for combinations of prognostic variables in models are shown for overall survival (OS) (A) and progression-free survival (PFS) (B). Combinations of prognostic variables contain the confirmed prognostic variables, as used in the reference model (age, sex, study medication, Eastern Cooperative Oncology Group [ECOG], log white blood cells [WBC], β2-microglobulin [β2- m], log thymidine kinase [TK], IGHV mutation status, del(11q), del(13q), del(17p), trisomy 12, TP53 mutation, NOTCH1 mutation, SF3B1 mutation) and gene expression profiling (GEP) variables. Prognostic GEP variables were selected in addition to (fixed model) or instead of (equally penalized model) the confirmed prognostic variables. In a separate approach prognostic GEP variables were selected in addition to (fixed model) or instead of (equally penalized model) non-genetic prognostic variables (only age, sex, study medication, ECOG, log WBC, log TK, β2-m). GEP variables selected in the fixed or equally penalized model largely overlap with the full prognostic gene signature (Online Supplementary Table S2), which is separately used in the “GEP data only” prediction error curve. Combination of prognostic variables selected in the equally penalized model performed highly similar to the model containing only confirmed prognostic variables. Strong overlap was found for prediction error curves represented by the red and blue solid lines.
Figure 2.
Figure 2.
Conditional Kaplan-Meier survival estimates illustrate the distribution for overall survival and progression-free survival within the different prediction models. Kaplan-Meier estimates were generated for the lowest, the median, and the highest observed values of the prognostic variable combinations. Kaplan-Meier estimates illustrate overall survival (OS) (A, C and E) and progression-free survival (PFS) (B, D and F) with regard to the “reference model” (confirmed prognostic variables only, A and B), the “equally penalized model” (confirmed prognostic variables and GEP equally penalized, C and D) and prognostic GEP signatures only (as represented in the Online Supplementary Table S2A and B) (E and F).
Figure 3.
Figure 3.
Association of RGS1, LDOC1 and L3MBTL4 with genetic variables. Boxplots showing distribution for log2 expression of genes selected for both overall survival (OS) and progressionfree survival (PFS), namely RGS1, LDOC1 and L3MBTL4. LDOC1 and L3MBTL4 show a bimodal distribution. Distribution of the three genes was not exclusively associated with distinct genetic variables.
Figure 4.
Figure 4.
Assessment of genes showing concordant or discordant expression with RGS1, LDOC1 and L3MBTL4. (A) Venn diagram illustrating overlaps for differentially expressed genes (fold-change [FC] >1.5; false discovery rate [FDR] <0.01) between patient samples with either high or low expression (upper vs. lower quartile) for RGS1, LDOC1 and L3MBTL4. (B) Heatmap showing clustered expression pattern (Pearson correlation and average linkage) of 12 genes found in all three gene specific signatures and heatmap showing expression pattern of 51 genes found in gene specific signatures of LDOC1 and L3MBTL4. (C) Scatter plots for ZAP70 expression with regard to groups showing high and low LDOC1 and L3MBTL4 expression (upper vs. lower quartile).
Figure 5.
Figure 5.
Combined status of LDOC1 and L3MBTL4 is correlated with IGHV sequence homology and identifies cases with “discordant” clinical course. The figure highlights the correlation between expression levels of LDOC1 (x-axis), L3MBTL4 (y-axis) and the immunoglobulin heavy chain variable (IGHV) gene sequence homology (color coded). Cases with IGHV sequence homology <98% are indicated in blue, cases with IGHV sequence homology ≥98% are indicated in red. LDOC1 and L3MBTL4 expression identifies “discordant” cases with mutated IGHV but poor clinical course (high expression of LDOC1 and/or L3MBTL4) and vice versa.

References

    1. Fischer K, Bahlo J, Fink AM, et al. . Long-term remissions after FCR chemoimmunotherapy in previously untreated patients with CLL: updated results of the CLL8 trial. Blood. 2016;127(2):208-215.
    1. Keating MJ, O`Brien S, Albitar M, et al. . Early results of a chemoimmunotherapy regimen of fludarabine, cyclophosphamide, and rituximab as initial therapy for chronic lymphocytic leukemia. J Clin Oncol. 2005;23(18):4079-4088.
    1. Rossi D, Rasi S, Fabbri G, et al. . Mutations of NOTCH1 are an independent predictor of survival in chronic lymphocytic leukemia. Blood. 2012;119(2):521-529.
    1. Döhner H, Stilgenbauer S, Benner A, et al. . Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343(26):1910-1916.
    1. Stilgenbauer S, Schnaiter A, Paschka P, et al. . Gene mutations and treatment outcome in chronic lymphocytic leukemia: results from the CLL8 trial. Blood. 2014;123(21):3247-3254.
    1. Damle RN, Wasil T, Fais F, et al. . Ig V gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood. 1999;94(6):1840-1847.
    1. Hamblin TJ, Davis Z, Gardiner A, Oscier DG, Stevenson FK. Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood. 1999;94(6):1848-1854.
    1. Woyach JA, Ruppert AS, Heerema NA, et al. . Ibrutinib regimens versus chemoimmunotherapy in older patients with untreated CLL. N Engl J Med. 2018;379(26):2517-2528.
    1. Moreno C, Greil R, Demirkan F, et al. . Ibrutinib plus obinutuzumab versus chlorambucil plus obinutuzumab in first-line treatment of chronic lymphocytic leukaemia (iLLUMINATE): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20(1):43-56.
    1. Roberts AW, Davids MS, Pagel JM, et al. . Targeting BCL2 with Venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374(4):311-322.
    1. Stilgenbauer S, Eichhorst B, Schetelig J, et al. . Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol. 2016;17(6):768-778.
    1. Byrd JC, Furman RR, Coutre SE, et al. . Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369(1):32-42.
    1. Furman RR, Sharman JP, Coutre SE, et al. . Idelalisib and Rituximab in relapsed chronic lymphocytic leukemia. N Engl J Med. 2014;370(11):997-1007.
    1. Shanafelt TD, Wang V, Kay NE, et al. . A randomized phase III study of ibrutinib (PCI- 32765)-based therapy vs. standard fludarabine, cyclophosphamide, and rituximab (FCR) chemoimmunotherapy in untreated younger patients with chronic lymphocytic leukemia (CLL): a trial of the ECOG-ACRIN cancer research group (E1912). Blood. 2018;132(Supplement 1):LBA-4.
    1. Bengtsson H, Simpson K, Bullard J, Hansen K. aroma.affymetrix: A generic framework in R for analyzing small to very large Affymetrix data sets in bounded memory. Tech Reports. 2008;745:1-9.
    1. van Buuren S, Groothuis-Oudshoorn K. MICE: multivariate imputation by chained equations in R. J Stat Software. 2011;45(3).
    1. Fan J, Li R. Variable selection via nonconcave penalized likelihood and its oracle properties. J Am Stat Assoc. 2001;96:1348-1360.
    1. Willi Sauerbrei, Buchholz A, Boulesteix AL, Binder H. On stability issues in deriving multivariable regression models. Biom J. 2015;57(4):531-555.
    1. Mogensen UB, Ishwaran H, Gerds TA. Evaluating random forests for survival analysis using prediction error curves. J Stat Softw. 2012;50(11):1-23.
    1. Beran R. Nonparametric regression with randomly censored survival data. Tech Report. 1981 University of California, Berkeley.
    1. Gerds TA. Prodlim: Product-limit estimation for censored event history analysis 2014. URL . R Packag. version 1, 460 (2016).
    1. Herold T, Jurinovic V, Metzeler KH, et al. . An eight-gene expression signature for the prediction of survival and time to treatment in chronic lymphocytic leukemia. Leukemia. 2011;25(10):1639-1645.
    1. Duzkale H, Schweighofer CD, Coombes KR, et al. . LDOC1 mRNA is differentially expressed in chronic lymphocytic leukemia and predicts overall survival in untreated patients. Blood. 2011;117(15):4076-4084.
    1. Morabito F, Cutrona G, Mosca L, et al. . Surrogate molecular markers for IGHV mutational status in chronic lymphocytic leukemia for predicting time to first treatment. Leuk Res. 2015;39(8):840-845.
    1. Rosenwald A, Alizadeh AA, Widhopf G, et al. . Relation of gene expression phenotype to immunoglobulin mutation genotype in B cell chronic lymphocytic leukemia. J Exp Med. 2001;194(11):1639-1647.
    1. Rassenti LZ, Huynh L, Toy TL, et al. . ZAP-70 compared with immunoglobulin heavychain gene mutation status as a predictor of disease progression in chronic lymphocytic leukemia. N Engl J Med. 2004;351(9):893-901.
    1. Klein U, Tu Y, Stolovitzky GA, et al. . Gene expression profiling of B cell chronic lymphocytic leukemia reveals a homogeneous phenotype related to memory B cells. J Exp Med. 2001;194(11):1625-1638.
    1. International CLL-IPI working group. An international prognostic index for patients with chronic lymphocytic leukaemia (CLLIPI): a meta-analysis of individual patient data. Lancet Oncol. 2016;17(6):779-790.
    1. Kulis M, Heath S, Bibikova M, et al. . Epigenomic analysis detects widespread gene-body DNA hypomethylation in chronic lymphocytic leukemia. Nat Genet. 2012;44(11):1236-1242.
    1. Oakes CC, Seifert M, Assenov Y, et al. . DNA methylation dynamics during B cell maturation underlie a continuum of disease phenotypes in chronic lymphocytic leukemia. Nat Genet. 2016;48(3):253-264.
    1. Wojdacz TK, Amarasinghe HE, Kadalayil L, et al. . Clinical significance of DNA methylation in chronic lymphocytic leukemia patients: results from 3 UK clinical trials. Blood Adv. 2019;3(16):2474-2481.
    1. Dvinge H, Ries RE, Ilagan JO, Stirewalt DL, Meshinchi S, Bradley RK. Sample processing obscures cancer-specific alterations in leukemic transcriptomes. Proc Natl Acad Sc. USA. 2014;111(47):16802-16807.
    1. Chen Q, Jain N, Ayer T, et al. . Economic burden of chronic lymphocytic leukemia in the era of oral targeted therapies in the United States. J Clin Oncol. 2017;35(2):166-174.
    1. Zhang W, Yu Y, Hertwig F, et al. . Comparison of RNA-seq and microarray-based models for clinical endpoint prediction. Genome Biol. 2015;16(1):133.

Source: PubMed

3
Suscribir