Next-generation sequencing-based monitoring of circulating tumor DNA reveals clonotypic heterogeneity in untreated PTCL

Milos D Miljkovic, Christopher Melani, Stefania Pittaluga, Rahul Lakhotia, Nicole Lucas, Allison Jacob, Erik Yusko, Elaine S Jaffe, Wyndham H Wilson, Mark Roschewski, Milos D Miljkovic, Christopher Melani, Stefania Pittaluga, Rahul Lakhotia, Nicole Lucas, Allison Jacob, Erik Yusko, Elaine S Jaffe, Wyndham H Wilson, Mark Roschewski

Abstract

Peripheral T-cell lymphomas (PTCLs) have marked biologic and clinical heterogeneity, which confounds treatment decisions. Advances in circulating tumor DNA (ctDNA) assays using next-generation sequencing (NGS) have improved the detection of molecular relapse and driver mutations in diffuse large B-cell lymphoma and show the potential utility of ctDNA across lymphomas. We investigated NGS-based monitoring of T-cell receptor (TCR) sequences in patients with PTCL undergoing frontline treatment. Of 45 patients, 34 (76%) had tumor-specific clonotypes of the TCRβ or TCRγ genes identified, which included 18 (86%) from baseline tissue and 16 (67%) from baseline serum. Twenty-five (74%) patients had both TCRβ and TCRγ clonotypes, 23 (68%) had more than 1 TCRγ clonotype, and 4 (9%) had multiple TCRβ or TCRγ clonotypes, demonstrating significant intrapatient clonotypic heterogeneity. Among 24 patients with available serial serum samples during treatment, 9 (38%) cleared ctDNA after 2 cycles of therapy, and 11 (46%) had detectable ctDNA at the end of treatment. Patients with detectable ctDNA after therapy showed a trend toward worse survival. Notably, 2 patients with persistently detectable ctDNA after therapy remained in remission with 10 years of follow-up. Clonotypic heterogeneity in tumors and persistence, despite long-term remission, suggests variability in oncological potential. This trial was registered at www.clinicaltrials.gov as #NCT00001337.

© 2021 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Patients and calibration. Fifty-four patients with untreated PTCL enrolled in the treatment study and were screened to determine whether baseline FFPE and/or serum samples were available. Thirty-four patients had a dominant T-cell receptor rearrangement identified from FFPE or serum samples, and they comprised the study cohort for the correlative analysis of circulating tumor DNA.
Figure 2.
Figure 2.
Identification of dominant T-cell receptor clonotypes from stored tissue biopsy specimens. (A) The number of dominant clonotypes of TCRγ (mustard) and TCRβ (lavender) identified in both baseline FFPE and serum samples from 18 patients who had at least 1 dominant TCR clonotype identified from baseline FFPE tissue. Of the 18, 16 had matched serum samples, with a dominant clonotype detected in 15. Thirteen dominant TCR clonotypes (lime green) across 7 of those 15 patients were identified in serum that were not identified in FFPE samples. (B) The number of dominant TCRγ (mustard) and TCRβ (lavender) clonotypes detected in baseline serum in 16 patients without available baseline FFPE samples. (C) Tissue source of all dominant TCR clonotypes detected exclusively in FFPE and serum samples, as well as those detected in both tissue sources. (D) The median number and range of clonotypes identified in baseline tissue samples across histologic subtypes of PTCL. ALK− ALCL, ALK− negative ALCL; EATL, enteropathy associated T-cell lymphoma; HSTCL-gd, hepatosplenic T-cell lymphoma gamma delta; PCGDTCL, primary cutaneous gamma delta T-cell lymphoma; PTCL-NOS-L, lymphoepithelioid cell variant of PTCL-NOS; PTCL-TFH, PTCL-T-follicular helper phenotype; SPLTCL, subcutaneous panniculitislike T-cell lymphoma.
Figure 3.
Figure 3.
Correlation of ctDNA values with other prognostic parameters. (A) Baseline levels of ctDNA) across IPI categories. (B) Baseline levels of ctDNA correlated with pretreatment serum LDH. ρ, Spearman’s rank correlation coefficient.
Figure 4.
Figure 4.
Association between dichotomized ctDNA level and survival. (A) EFS in patients with baseline levels of ctDNA above the median level compared with patients with levels below the median. (B) OS in patients with baseline levels of ctDNA above the median level compared with patients with levels below the median.
Figure 5.
Figure 5.
ctDNA kinetics during therapy and clinical outcomes (A) The median and range of quantitative ctDNA levels after each cycle of therapy until treatment is complete. Levels of ctDNA in patients with ALK+ ALCL are depicted with green dots, and levels in other histologic subtypes of PTCL are depicted with black dots. (B) EFS based on presence or absence of ctDNA as a marker of MRD, drawn before day 1 of cycle 3. (C) OS based on the presence or absence of ctDNA as a marker of MRD drawn before day 1 of cycle 3. (D) EFS based on the presence or absence of ctDNA as a marker of MRD at the EoT. (E) OS based on presence or absence of ctDNA as a marker of MRD at EoT. C, cycle.
Figure 6.
Figure 6.
ctDNA as surveillance monitoring in clinical progressors. The results of ctDNA detected in stored serum samples collected after therapy compared with conventional monitoring for clinical relapse with periodic clinic visits and regular CT scans. (A) Serum samples collected after therapy in 8 patients who achieved CR, underwent surveillance, and then clinically progressed. The assigned patient numbers are to the left. Histology: patient 9, PTCL-NOS; patients 180, 291, and 317, AITL; patients 190 and 200, ALK+ ALCL; and patients 198 and 274, PTCL-TFH. (B) The pattern of fluctuation of quantitative ctDNA levels of patients 9 and 198 with persistently detectable ctDNA before clinical relapse. The x-axis represents time since the on-study date in months and is different for each graph. The y-axis represents the log10 of the quantity of each dominant clonotype (in clonotypes per milliliter). Each colored line represents a different clonotype. The horizontal gray line represents the median level of baseline ctDNA. The dashed vertical line represents EoT, the red vertical line represents time of progression . The green horizontal bar represents time in clinical remission, and the red horizontal bar represents time after progression.
Figure 7.
Figure 7.
Role of ctDNA in surveillance monitoring in nonprogressors. (A) Serum samples collected after therapy in 8 patients who achieved a complete response and underwent surveillance but did not progress clinically. The assigned patient numbers are on the left. Histology: patients 1, 8, and 316, ALK− ALCL; patients 3, 14, and 221, ALK+ALCL; patient 12, PTCL-NOS; and patient 15, AITL. (B) The pattern of fluctuation of quantitative ctDNA levels of patients 1 and 15 with multiple time points of detectable ctDNA after therapy but no overt clinical relapse. The x-axis represents time since the on-study date in months and is different for each graph. The y-axis represents the log10 of the quantity of each dominant clonotype (in clonotypes per milliliter). Each colored line represents a different clonotype. The horizontal gray line represents the median level of baseline ctDNA. The dashed vertical line represents EoT, the red vertical line represents time of progression, and the black vertical line represents end of follow-up. The green horizontal bar represents time in clinical remission.

References

    1. Vose J, Armitage J, Weisenburger D; International T-Cell Lymphoma Project .International peripheral T-cell and natural killer/T-cell lymphoma study: pathology findings and clinical outcomes. J Clin Oncol. 2008;26(25):4124-4130.
    1. Schmitz N, Trümper L, Ziepert M, et al. . Treatment and prognosis of mature T-cell and NK-cell lymphoma: an analysis of patients with T-cell lymphoma treated in studies of the German High-Grade Non-Hodgkin Lymphoma Study Group. Blood. 2010;116(18):3418-3425.
    1. Dunleavy K, Pittaluga S, Shovlin M, et al. . Phase II trial of dose-adjusted EPOCH in untreated systemic anaplastic large cell lymphoma. Haematologica. 2016;101(1):e27-e29.
    1. Maeda Y, Nishimori H, Yoshida I, et al. . Dose-adjusted EPOCH chemotherapy for untreated peripheral T-cell lymphomas: a multicenter phase II trial of West-JHOG PTCL0707. Blood. 2017;102(12):2097-2103.
    1. d’Amore F, Relander T, Lauritzsen GF, et al. . Up-front autologous stem-cell transplantation in peripheral T-cell lymphoma: NLG-T-01. J Clin Oncol. 2012;30(25):3093-3099.
    1. Rohlfing S, Dietrich S, Witzens-Harig M, et al. . The impact of stem cell transplantation on the natural course of peripheral T-cell lymphoma: a real-world experience. Ann Hematol. 2018;97(7):1241-1250.
    1. Janikova A, Chloupkova R, Campr V, et al. . First-line therapy for T cell lymphomas: a retrospective population-based analysis of 906 T cell lymphoma patients. Ann Hematol. 2019;98(8):1961-1972.
    1. Kuzich JA, Hutchison AP, Lim KJC, et al. . Prognostic factors and the impact of frontline therapy in peripheral T-cell lymphoma: 10 years of ‘real-world’ experience from Western Australia. Leuk Lymphoma. 2019; 60(14):3417-3425.
    1. Roerden M, Walz JS, Müller MR, et al. . The role of autologous stem cell transplantation in peripheral T cell lymphoma: a long-term follow-up single-center experience. J Cancer Res Clin Oncol. 2019;145(10):2595-2604.
    1. Berg H, Otteson GE, Corley H, et al. . Flow cytometric evaluation of TRBC1 expression in tissue specimens and body fluids is a novel and specific method for assessment of T-cell clonality and diagnosis of T-cell neoplasms. Cytometry B Clin Cytom. 2021;100(3):361–369.
    1. Shi M, Jevremovic D, Otteson GE, Timm MM, Olteanu H, Horna P.. Single Antibody Detection of T-Cell Receptor αβ Clonality by Flow Cytometry Rapidly Identifies Mature T-Cell Neoplasms and Monotypic Small CD8-Positive Subsets of Uncertain Significance. Cytometry B Clin Cytom. 2020; 98(1):99-107.
    1. Schützinger C, Esterbauer H, Hron G, et al. . Prognostic value of T-cell receptor gamma rearrangement in peripheral blood or bone marrow of patients with peripheral T-cell lymphomas. Leuk Lymphoma. 2008;49(2):237-246.
    1. de Masson A, O’Malley JT, Elco CP, et al. . High-throughput sequencing of the T cell receptor β gene identifies aggressive early-stage mycosis fungoides. Sci Transl Med. 2018;10(440):eaar5894.
    1. Rea B, Haun P, Emerson R, et al. . Role of high-throughput sequencing in the diagnosis of cutaneous T-cell lymphoma. J Clin Pathol. 2018; 71(9):814-820.
    1. Roschewski M, Dunleavy K, Pittaluga S, et al. . Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: a correlative biomarker study. Lancet Oncol. 2015;16(5):541-549.
    1. Kurtz DM, Green MR, Bratman SV, et al. . Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679-3687.
    1. Scherer F, Kurtz DM, Newman AM, et al. . Distinct biological subtypes and patterns of genome evolution in lymphoma revealed by circulating tumor DNA. Sci Transl Med. 2016;8(364):364ra155.
    1. Kurtz DM, Scherer F, Jin MC, et al. . Circulating Tumor DNA Measurements As Early Outcome Predictors in Diffuse Large B-Cell Lymphoma. J Clin Oncol. 2018;36(28):2845-2853.
    1. Kurtz DM, Esfahani MS, Scherer F, et al. . Dynamic Risk Profiling Using Serial Tumor Biomarkers for Personalized Outcome Prediction. Cell. 2019; 178(3):699-713.e19.
    1. Swerdlow SH, Campo E, Pileri SA, et al. . The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016; 127(20):2375-2390.
    1. Wilson WH, Grossbard ML, Pittaluga S, et al. . Dose-adjusted EPOCH chemotherapy for untreated large B-cell lymphomas: a pharmacodynamic approach with high efficacy. Blood. 2002;99(8):2685-2693.
    1. Cheson BD, Pfistner B, Juweid ME, et al. ; International Harmonization Project on Lymphoma . Revised response criteria for malignant lymphoma. J Clin Oncol. 2007;25(5):579-586.
    1. Juweid ME, Wiseman GA, Vose JM, et al. . Response assessment of aggressive non-Hodgkin’s lymphoma by integrated International Workshop Criteria and fluorine-18-fluorodeoxyglucose positron emission tomography. J Clin Oncol. 2005;23(21):4652-4661.
    1. Weng W-K, Armstrong R, Arai S, Desmarais C, Hoppe R, Kim YH.. Minimal Residual Disease Monitoring with High-Throughput Sequencing of T Cell Receptors in Cutaneous T Cell Lymphoma. Sci Transl Med. 2013;5(214):214ra171.
    1. Carlson CS, Emerson RO, Sherwood AM, et al. . Using synthetic templates to design an unbiased multiplex PCR assay. Nat Commun. 2013;4:2680.
    1. Wu D, Sherwood A, Fromm JR, et al. . High-Throughput Sequencing Detects Minimal Residual Disease in Acute T Lymphoblastic Leukemia. Sci Transl Med. 2012;4(134):134ra163.
    1. El-Asmar J, Reljic T, Ayala E, et al. . Efficacy of High-Dose Therapy and Autologous Hematopoietic Cell Transplantation in Peripheral T Cell Lymphomas as Front-Line Consolidation or in the Relapsed/Refractory Setting: A Systematic Review/Meta-Analysis. Biol Blood Marrow Transplant. 2016;22(5):802-814.
    1. Fossard G, Broussais F, Coelho I, et al. . Role of up-front autologous stem-cell transplantation in peripheral T-cell lymphoma for patients in response after induction: an analysis of patients from LYSA centers. Ann Oncol. 2018;29(3):715-723.
    1. Kanakry JA, Kasamon YL, Gocke CD, et al. . Outcomes of related donor HLA-identical or HLA-haploidentical allogeneic blood or marrow transplantation for peripheral T cell lymphoma. Biol Blood Marrow Transplant. 2013;19(4):602-606.
    1. Blüthmann H, Kisielow P, Uematsu Y, et al. . T-cell-specific deletion of T-cell receptor transgenes allows functional rearrangement of endogenous alpha- and beta-genes. Nature. 1988;334(6178):156-159.
    1. Yassai MB, Naumov YN, Naumova EN, Gorski J.. A clonotype nomenclature for T cell receptors. Immunogenetics. 2009;61(7):493-502.
    1. Kanavaros P, Farcet JP, Gaulard P, et al. . Recombinative events of the T cell antigen receptor delta gene in peripheral T cell lymphomas. J Clin Invest. 1991;87(2):666-672.
    1. Gong Q, Wang C, Zhang W, et al. . Assessment of T-cell receptor repertoire and clonal expansion in peripheral T-cell lymphoma using RNA-seq data. Sci Rep. 2017;7(1):11301.
    1. Hamrouni A, Fogh H, Zak Z, Ødum N, Gniadecki R.. Clonotypic diversity of the T-cell receptor corroborates the immature precursor origin of cutaneous T-cell lymphoma. Clin Cancer Res. 2019;25(10):3104-3114.
    1. Iyer A, Hennessey D, O’Keefe S, et al. . Clonotypic heterogeneity in cutaneous T-cell lymphoma (mycosis fungoides) revealed by comprehensive whole-exome sequencing. Blood Adv. 2019;3(7):1175-1184.
    1. Farmanbar A, Kneller R, Firouzi S.. RNA sequencing identifies clonal structure of T-cell repertoires in patients with adult T-cell leukemia/lymphoma. NPJ Genom Med. 2019;4(1):10.
    1. Rauch DA, Conlon KC, Janakiram M, et al. . Rapid progression of adult T-cell leukemia/lymphoma as tumor infiltrating Treg cells after PD-1 blockade. Blood. 2019; 134(17):1406-1414.
    1. Ottolini B, Nawaz N, Trethewey CS, et al. . Multiple mutations at exon 2 of RHOA detected in plasma from patients with peripheral T-cell lymphoma. Blood Adv. 2020;4(11):2392-2403.
    1. Schatz JH, Horwitz SM, Teruya-Feldstein J, et al. . Targeted mutational profiling of peripheral T-cell lymphoma not otherwise specified highlights new mechanisms in a heterogeneous pathogenesis. Leukemia. 2015;29(1):237-241.
    1. Watatani Y, Sato Y, Miyoshi H, et al. . Molecular heterogeneity in peripheral T-cell lymphoma, not otherwise specified revealed by comprehensive genetic profiling. Leukemia. 2019;33(12):2867-2883.
    1. Theodorou I, Raphaël M, Bigorgne C, et al. . Recombination pattern of the TCR gamma locus in human peripheral T-cell lymphomas. J Pathol. 1994; 174(4):233-242.
    1. Tan BT, Warnke RA, Arber DA.. The frequency of B- and T-cell gene rearrangements and epstein-barr virus in T-cell lymphomas: a comparison between angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, unspecified with and without associated B-cell proliferations. J Mol Diagn. 2006;8(4):466-475, quiz 527.

Source: PubMed

3
購読する