Clonal dynamics towards the development of venetoclax resistance in chronic lymphocytic leukemia

Carmen D Herling, Nima Abedpour, Jonathan Weiss, Anna Schmitt, Ron Daniel Jachimowicz, Olaf Merkel, Maria Cartolano, Sebastian Oberbeck, Petra Mayer, Valeska Berg, Daniel Thomalla, Nadine Kutsch, Marius Stiefelhagen, Paula Cramer, Clemens-Martin Wendtner, Thorsten Persigehl, Andreas Saleh, Janine Altmüller, Peter Nürnberg, Christian Pallasch, Viktor Achter, Ulrich Lang, Barbara Eichhorst, Roberta Castiglione, Stephan C Schäfer, Reinhard Büttner, Karl-Anton Kreuzer, Hans Christian Reinhardt, Michael Hallek, Lukas P Frenzel, Martin Peifer, Carmen D Herling, Nima Abedpour, Jonathan Weiss, Anna Schmitt, Ron Daniel Jachimowicz, Olaf Merkel, Maria Cartolano, Sebastian Oberbeck, Petra Mayer, Valeska Berg, Daniel Thomalla, Nadine Kutsch, Marius Stiefelhagen, Paula Cramer, Clemens-Martin Wendtner, Thorsten Persigehl, Andreas Saleh, Janine Altmüller, Peter Nürnberg, Christian Pallasch, Viktor Achter, Ulrich Lang, Barbara Eichhorst, Roberta Castiglione, Stephan C Schäfer, Reinhard Büttner, Karl-Anton Kreuzer, Hans Christian Reinhardt, Michael Hallek, Lukas P Frenzel, Martin Peifer

Abstract

Deciphering the evolution of cancer cells under therapeutic pressure is a crucial step to understand the mechanisms that lead to treatment resistance. To this end, we analyzed whole-exome sequencing data of eight chronic lymphocytic leukemia (CLL) patients that developed resistance upon BCL2-inhibition by venetoclax. Here, we report recurrent mutations in BTG1 (2 patients) and homozygous deletions affecting CDKN2A/B (3 patients) that developed during treatment, as well as a mutation in BRAF and a high-level focal amplification of CD274 (PD-L1) that might pinpoint molecular aberrations offering structures for further therapeutic interventions.

Conflict of interest statement

C.D.H, C.-M.W., B.E., K.-A.K., M.H., and L.P.F. received research funding from Hofmann-La Roche. Research support was also provided by AbbVie to P.C., C.-M.W., B.E., K.-A.K., and M.H. P.C., C.-M.W., B.E., K.-A.K., H.C.R., M.H., and L.P.F. obtained consulting and/or speaker’s honoraria from AbbVie. P.C., C.-M.W., B.E., K.-A.K., and M.H. received consulting and/or speaker’s honoraria from Hofmann-La Roche. AbbVie provided travel support to P.C., N.K., and L.P.F. The remaining authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
Patient and their related matched pre-treatment and relapse samples characteristics. a Absolute lymphocyte counts and lymphadenopathy of the patients during venetoclax therapy. Day zero marks the start of the venetoclax treatment. Green lines show the time points of sample collection. Computer tomography (CT) scans for staging were performed at the time points marked by red lines. b Results from whole-exome sequencing are shown, including: number of somatic mutations, sample ploidy, percent of the genome undergoing copy number alterations (blue for losses and red for gains), and cancer-related gene mutations with pronounced clonal dynamics during therapy. Genomic alterations are annotated according to the color panel below the image. Sample type/compartment and the status if a patient has undergone a Richter’s transformation are additionally indicated. Pre-treatment samples (T0) are shown in red. c Giemsa, Ki67, CD3, and CD274 stains from lymph node material of patient C811 after relapse from venetoclax. High protein levels of CD274 are consistent with the genomic amplification of the locus containing CD274. Scale bar, 100 µm
Fig. 2
Fig. 2
Heterogeneous clonal evolutions under venetoclax therapy. Phylogenetic trees at the left side of each panel demonstrate the clonal evolution of the reconstructed cell populations for each patient. Highlighted mutations that occurred during tumor evolution are present in all descendent clones. Therefore, mutations in the most common ancestor population (C0) are present in all analyzed samples at a clonal level. The second type trees (right-bottom of each panel) demonstrate the phylogenetic relations of the matched pre-treatment and relapse samples from a patient, as commonly used in other cancer evolution studies,. Clonal composition of the samples (top-right of each panel) provides a link between both types of phylogenetic trees. We inferred diverse evolutionary paths across the patients: a linear evolution (C789), b branching evolution into three lineages (C577), c divergent evolution of two branches (C548), and d convergent evolution (C586). Pre-treatment sample names are displayed in red. Notable gene alterations are shown in the context of the ancestral relation of the clones
Fig. 3
Fig. 3
Overexpression of oncogenic BRAF in the OCI-LY19 cell line. a Western blot analysis of BRAF and MCL1 in the BRAFV600E overexpressing OCI-LY19 cell line vs. its empty vector control. b Growth inhibition of BRAFV600E transfected OCI-LY19 cells and the empty vector control is shown as a function of the concentration of venetoclax

References

    1. Binet JL, et al. A clinical staging system for chronic lymphocytic leukemia: prognostic significance. Cancer. 1977;40:855–864. doi: 10.1002/1097-0142(197708)40:2<855::AID-CNCR2820400239>;2-1.
    1. Dohner H, et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N. Engl. J. Med. 2000;343:1910–1916. doi: 10.1056/NEJM200012283432602.
    1. Landau DA, et al. Mutations driving CLL and their evolution in progression and relapse. Nature. 2015;526:525–530. doi: 10.1038/nature15395.
    1. Landau DA, et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell. 2013;152:714–726. doi: 10.1016/j.cell.2013.01.019.
    1. Puente XS, et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature. 2011;475:101–105. doi: 10.1038/nature10113.
    1. Quesada V, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat. Genet. 2011;44:47–52. doi: 10.1038/ng.1032.
    1. Schuh A, et al. Monitoring chronic lymphocytic leukemia progression by whole genome sequencing reveals heterogeneous clonal evolution patterns. Blood. 2012;120:4191–4196. doi: 10.1182/blood-2012-05-433540.
    1. Puente XS, et al. Non-coding recurrent mutations in chronic lymphocytic leukaemia. Nature. 2015;526:519–524. doi: 10.1038/nature14666.
    1. Woyach JA, et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N. Engl. J. Med. 2014;370:2286–2294. doi: 10.1056/NEJMoa1400029.
    1. de Bruin EC, et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014;346:251–256. doi: 10.1126/science.1253462.
    1. Jamal-Hanjani M, et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2109–2121. doi: 10.1056/NEJMoa1616288.
    1. Nik-Zainal S, et al. The life history of 21 breast cancers. Cell. 2012;149:994–1007. doi: 10.1016/j.cell.2012.04.023.
    1. Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014;346:256–259. doi: 10.1126/science.1256930.
    1. Burger JA, et al. Clonal evolution in patients with chronic lymphocytic leukaemia developing resistance to BTK inhibition. Nat. Commun. 2016;7:11589. doi: 10.1038/ncomms11589.
    1. Roberts AW, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 2016;374:311–322. doi: 10.1056/NEJMoa1513257.
    1. Stilgenbauer S, et al. Venetoclax in relapsed or refractory chronic lymphocytic leukaemia with 17p deletion: a multicentre, open-label, phase 2 study. Lancet Oncol. 2016;17:768–778. doi: 10.1016/S1470-2045(16)30019-5.
    1. Bowyer SE, et al. Activity of trametinib in K601E and L597Q BRAF mutation-positive metastatic melanoma. Melanoma Res. 2014;24:504–508. doi: 10.1097/CMR.0000000000000099.
    1. Brahmer JR, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012;366:2455–2465. doi: 10.1056/NEJMoa1200694.
    1. Budczies J, et al. Pan-cancer analysis of copy number changes in programmed death-ligand 1 (PD-L1, CD274)—associations with gene expression, mutational load, and survival. Genes. Chromosomes Cancer. 2016;55:626–639. doi: 10.1002/gcc.22365.
    1. George J, et al. Genomic amplification of CD274 (PD-L1) in small-cell lung cancer. Clin. Cancer Res. 2017;23:1220–1226. doi: 10.1158/1078-0432.CCR-16-1069.
    1. Kuo ML, et al. Arf induces p53-dependent and -independent antiproliferative genes. Cancer Res. 2003;63:1046–1053.
    1. Nahta R, et al. B cell translocation gene 1 contributes to antisense Bcl-2-mediated apoptosis in breast cancer cells. Mol. Cancer Ther. 2006;5:1593–1601. doi: 10.1158/1535-7163.MCT-06-0133.
    1. Chigrinova E, et al. Two main genetic pathways lead to the transformation of chronic lymphocytic leukemia to Richter syndrome. Blood. 2013;122:2673–2682. doi: 10.1182/blood-2013-03-489518.
    1. Fabbri G, et al. Genetic lesions associated with chronic lymphocytic leukemia transformation to Richter syndrome. J. Exp. Med. 2013;210:2273–2288. doi: 10.1084/jem.20131448.
    1. Quentmeier H, et al. Subclones in B-lymphoma cell lines: isogenic models for the study of gene regulation. Oncotarget. 2016;7:63456–63465. doi: 10.18632/oncotarget.11524.
    1. Hertlein E, et al. Characterization of a new chronic lymphocytic leukemia cell line for mechanistic in vitro and in vivo studies relevant to disease. PLoS ONE. 2013;8:e76607. doi: 10.1371/journal.pone.0076607.
    1. Hallek M, 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;111:5446–5456. doi: 10.1182/blood-2007-06-093906.
    1. George J, et al. Comprehensive genomic profiles of small cell lung cancer. Nature. 2015;524:47–53. doi: 10.1038/nature14664.
    1. Peifer M, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat. Genet. 2012;44:1104–1110. doi: 10.1038/ng.2396.
    1. Peifer M, et al. Telomerase activation by genomic rearrangements in high-risk neuroblastoma. Nature. 2015;526:700–704. doi: 10.1038/nature14980.
    1. Felsenstein J. PHYLIP - Phylogeny Inference Package (Version 3.2) Cladistics. 1989;5:164–166.
    1. Assenov Y, et al. Comprehensive analysis of DNA methylation data with RnBeads. Nat. Methods. 2014;11:1138–1140. doi: 10.1038/nmeth.3115.
    1. Teschendorff AE, et al. A beta-mixture quantile normalization method for correcting probe design bias in Illumina Infinium 450k DNA methylation data. Bioinformatics. 2013;29:189–196. doi: 10.1093/bioinformatics/bts680.
    1. Clinical Lung Cancer Genome CLCGP, Network Genomic NGM. A genomics-based classification of human lung tumors. Sci. Transl. Med. 2013;5:209ra153.
    1. Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods. 2014;11:783–784. doi: 10.1038/nmeth.3047.

Source: PubMed

3
Sottoscrivi