Targeted genotyping of circulating tumor DNA for classical Hodgkin lymphoma monitoring: a prospective study

Vincent Camus, Mathieu Viennot, Justine Lequesne, Pierre-Julien Viailly, Elodie Bohers, Lucile Bessi, Bénédicte Marcq, Pascaline Etancelin, Sydney Dubois, Jean-Michel Picquenot, Elena-Liana Veresezan, Marie Cornic, Lucie Burel, Justine Loret, Stéphanie Becker, Pierre Decazes, Pascal Lenain, Stéphane Lepretre, Emilie Lemasle, Hélène Lanic, Anne-Lise Ménard, Nathalie Contentin, Hervé Tilly, Aspasia Stamatoullas, Fabrice Jardin, Vincent Camus, Mathieu Viennot, Justine Lequesne, Pierre-Julien Viailly, Elodie Bohers, Lucile Bessi, Bénédicte Marcq, Pascaline Etancelin, Sydney Dubois, Jean-Michel Picquenot, Elena-Liana Veresezan, Marie Cornic, Lucie Burel, Justine Loret, Stéphanie Becker, Pierre Decazes, Pascal Lenain, Stéphane Lepretre, Emilie Lemasle, Hélène Lanic, Anne-Lise Ménard, Nathalie Contentin, Hervé Tilly, Aspasia Stamatoullas, Fabrice Jardin

Abstract

The relevance of circulating tumor DNA (ctDNA) analysis as a liquid biopsy and minimal residual disease tool in the management of classical Hodgkin Lymphoma (cHL) patients was demonstrated in retrospective settings and remains to be confirmed in a prospective setting. We developed a targeted Next-Generation sequencing (NGS) panel for fast analysis (AmpliSeq technology) of nine commonly mutated genes in biopies and ctDNA of cHL patients. We then conducted a prospective trial to assess ctDNA follow up at diagnosis and after 2 cycles of chemotherapy (C2). Sixty cHL patients treated by first line conventional chemotherapy (BEACOPPescalated [21.3%], ABVD/ABVD-like [73.5%] and other regimens [5.2%, for elderly patients] were assessed in this non-interventional study. Median age of the patients was 33.5 years (range 20-86). Variants were identified in 42 (70%) patients. Mutations of NFKBIE, TNFAIP3, STAT6, PTPN1, B2M, XPO1, ITPKB, GNA13 and SOCS1 were found in 13.3%, 31.7%, 23.3%, 5%, 33.3%, 10%, 23.3%, 13.3% and 50% of patients, respectively. ctDNA concentration and genotype are correlated with clinical characteristics and presentation. Regarding early therapeutic response, 45 patients (83%, NA=6) had a negative positron emission tomography (PET) after C2 (Deauville Score 1-3). Mean of DeltaSUVmax after C2 was -78.8%. We analyzed ctDNA after C2 for 54 patients (90%). ctDNA became rapidly undetectable in all cases after C2. Variant detection in ctDNA is suitable to depict the genetic features of cHL at diagnosis and may help to assess early treatment response, in association with PET. Clinical Trial reference: NCT02815137.

Figures

Figure 1.
Figure 1.
Number and types of somatic variants identified by gene and patient both in biopsy and circulating tumor DNA. (A) Heatmap representing somatic variants detected in circulating tumor DNA (ctDNA) in the mutated patients (the "Burden" track represents the mutational load [“burden”] of a patient based on the maximum/minimum number of mutations found in the complete cohort). (B) Unsupervised hierarchical clustering performed among the nine genes to represent the association of alterations (numbers within the clustering represent the number of observed associations between two genes).
Figure 2.
Figure 2.
Prevalence of somatic mutations detected in DNA extracted from the 31 available biopsies and 60 plasma samples of the patients at time of diagnosis. The "genomic DNA (gDNA) and/or circulating tumor DNA (ctDNA) " column corresponds to the patients considered mutated in the biopsy and/or the plasma samples.
Figure 3.
Figure 3.
Distribution of circulating tumor DNA median variant allelic frequency for the 41 mutated patients with positive plasma at diagnosis. ctDNA: circulating tumor DNA; VAF: variant allelic frequency
Figure 4.
Figure 4.
Longitudinal assessment of mutation abundance in plasma circulating tumor DNA upon treatment. Evolution of median circulating tumor DNA (ctDNA) variant allelic frequency (VAF) for each patient (with detectable ctDNA mutation at diagnosis) throughout treatment (at diagnosis [“diag”] n=41) and after two cycles (C2) of chemotherapy (“post C2”, n=31). UPN: universal patient identification number.
Figure 5.
Figure 5.
Example of possible application of liquid biopsy analysis. Patient (UPN38) presenting a positive positron emission tomography (PET) scan with residual fixation of a left supraclavicular lymph node (Deauville Scale 5) after two cycles (C2) of adriamycin (doxorubicin), bleomycin, vinblastine, dacarbazine (ABVD): the biopsy showed a reactive lymph node, indicating a false positive PET scan. Plasma circulating tumor DNA (ctDNA) sample was negative at time of the PET post C2 of ABVD with a clearance of all baseline mutations that were present at diagnosis. UPN: universal patient identification number.

References

    1. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease. N Engl J Med. 1998;339(21):1506-1514.
    1. Hutchings M, Loft A, Hansen M, et al. FDGPET after two cycles of chemotherapy predicts treatment failure and progression-free survival in Hodgkin lymphoma. Blood. 2006;107(1):52-59.
    1. Canellos GP, Rosenberg SA, Friedberg JW, Lister TA, Devita VT. Treatment of Hodgkin lymphoma: a 50-year perspective. J Clin Oncol. 2014;32(3):163-168.
    1. Schmitz R, Stanelle J, Hansmann M-L, Küppers R. Pathogenesis of classical and lymphocyte-predominant Hodgkin lymphoma. Annu Rev Pathol. 2009;4151-174.
    1. Diaz LA, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J Clin Oncol. 2014;32(6):579-586.
    1. Spina V, Bruscaggin A, Cuccaro A, et al. Circulating tumor DNA reveals genetics, clonal evolution, and residual disease in classical Hodgkin lymphoma. Blood. 2018; 131(22):2413-2425.
    1. Camus V, Stamatoullas A, Mareschal S, et al. Detection and prognostic value of recurrent exportin 1 mutations in tumor and cell-free circulating DNA of patients with classical Hodgkin lymphoma. Haematologica. 2016; 101(9):1094-1101.
    1. Bessi L, Viailly P-J, Bohers E, et al. Somatic mutations of cell-free circulating DNA detected by targeted next-generation sequencing and digital droplet PCR in classical Hodgkin lymphoma. Leuk Lymphoma. 2019;60(2):498-502.
    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. Bohers E, Viailly P-J, Becker S, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort. Blood Cancer J. 2018; 8(8):74.
    1. Salipante SJ, Adey A, Thomas A, et al. Recurrent somatic loss of TNFRSF14 in classical Hodgkin lymphoma. Genes Chromosomes Cancer. 2016;55(3):278-287.
    1. Vandenberghe P, Wlodarska I, Tousseyn T, et al. Non-invasive detection of genomic imbalances in Hodgkin/Reed-Sternberg cells in early and advanced stage Hodgkin’s lymphoma by sequencing of circulating cell-free DNA: a technical proof-of-principle study. Lancet Haematol. 2015;2(2):e55-e65.
    1. Reichel J, Chadburn A, Rubinstein PG, et al. Flow sorting and exome sequencing reveal the oncogenome of primary Hodgkin and Reed-Sternberg cells. Blood. 2015; 125(7):1061-1072.
    1. Mottok A, Renne C, Willenbrock K, Hansmann M-L, Brauninger A. Somatic hypermutation of SOCS1 in lymphocytepredominant Hodgkin lymphoma is accompanied by high JAK2 expression and activation of STAT6. Blood. 2007;110(9):3387-3390.
    1. Ritz O, Guiter C, Dorsch K, et al. STAT6 activity is regulated by SOCS-1 and modulates BCL-XL expression in primary mediastinal B-cell lymphoma. Leukemia. 2008; 22(11):2106-2110.
    1. Weniger MA, Melzner I, Menz CK, et al. Mutations of the tumor suppressor gene SOCS-1 in classical Hodgkin lymphoma are frequent and associated with nuclear phospho- STAT5 accumulation. Oncogene. 2006; 25(18):2679-2684.
    1. Bohers E, Viailly P-J, Becker S, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by cell-free DNA high-throughput targeted sequencing: analysis of a prospective cohort. Blood Cancer J. 2018; 8(8):74.
    1. Elshimali Y, Khaddour H, Sarkissyan M, Wu Y, Vadgama J. The clinical utilization of circulating cell free DNA (CCFDNA) in blood of cancer patients. Int J Mol Sci. 2013; 14(9):18925-18958.
    1. Delfau-Larue M-H, van der Gucht A, Dupuis J, et al. Total metabolic tumor volume, circulating tumor cells, cell-free DNA: distinct prognostic value in follicular lymphoma. Blood Adv. 2018;2(7):807-816.
    1. Kanakry JA, Li H, Gellert LL, et al. Plasma Epstein-Barr virus DNA predicts outcome in advanced Hodgkin lymphoma: correlative analysis from a large North American cooperative group trial. Blood. 2013; 121(18): 3547-3553.

Source: PubMed

3
Předplatit