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; doi: 10.1200/JCO.2018.78.5246. [Epub ahead of print].

Major advances in our understanding and treatment of diffuse large B-cell lymphoma (DLBCL) have come from genomic analysis of this disease. Several recent landmark studies have identified that adding pretreatment genomic information to established clinical prognostic tools in DLBCL such as the International Prognostic Index (IPI) score and interim positron emission tomography (PET) may greatly improve risk stratification.1-3  However, the connection between the use of this risk stratification to select patients for intensified therapy and improvement of patient outcome is not clear. To this end, a recent study by Dr. David M. Kurtz and colleagues rigorously set out to evaluate whether dynamic and serial measurements of circulating tumor DNA (ctDNA) from plasma of DLBCL patients could serve to predict outcome early during the course of treatment.

In this study, ctDNA from 217 patients with DLBCL or primary mediastinal B-cell lymphoma were assessed in a discovery and in two validation cohorts. The authors sequenced tumor and pretreatment cell-free DNA from plasma in all patients to identify somatic alterations for ctDNA quantitation and disease monitoring. They applied deep sequencing of genes for mutational genotyping and tracking of mutations before and during therapy for predicting event-free survival (EFS) at 24 months as well as overall survival (OS). Specifically, they performed cancer personalized profiling by deep sequencing (CAPP-Seq), a form of targeted deep next-generation sequencing invented by these authors.4  Using this method, 99 percent (215 of 217) of patients had at least one tumor-specific alteration detected for monitoring, and 98 percent (212 of 217) had ctDNA prior to therapy. Pretreatment ctDNA was significantly correlated with IPI, total metabolic tumor volume (TMTV), EFS, and OS in patients receiving frontline therapy. Moreover, pretreatment ctDNA was also associated with OS for patients receiving salvage therapy. Finally, in multivariate analysis, pretreatment ctDNA levels remained prognostically important when controlling for IPI, molecular subtype, and TMTV. Overall, these data indicate that ctDNA burden may itself have prognostic value in DLBCL.

One clear benefit of the use of ctDNA for disease monitoring is the ease with which repeated sampling of disease can be performed. Given this, the authors then investigated whether dynamic changes in ctDNA levels might also predict disease response. They densely sampled plasma from the discovery cohort of 14 patients and noted that achieving 1) a two-log (i.e., 100-fold) decrease in ctDNA between pretreatment samples and end of cycle 1 of therapy, or 2) a 2.5-log drop in ctDNA between pretreatment samples and end of cycle 3 of therapy, clearly separated patients who achieved a complete response from others. These same thresholds (defined by the authors as “early molecular response” [EMR] or “major molecular response” [MMR], depending on whether it was assessed at the end of cycle 1 or end of cycle 3, respectively) were confirmed in the two validation cohorts. Moreover, they identified that the line of therapy (frontline vs. salvage) or the type of therapy (R-CHOP [rituximab-cyclophosphamide, doxorubicine, vincristine, and prednisolone] vs. EPOCH-R [etoposide phosphate, prednisone, vincristine sulfate, cyclophosphamide, doxorubicin hydrochloride-rituximab]) did not influence these cut-offs in ctDNA metrics. Finally, EMR and MMR were also predictive of EFS and OS in patients receiving frontline therapy.

This study raises several points for future consideration. First, it begs for standardization and evaluation of widely available assays for molecular monitoring in DLBCL. As mentioned earlier, this study used CAPP-Seq, but alternative, commercially available methods for molecular monitoring in B-cell malignancies, including sequencing of immunoglobulin genes, should also be considered. Finally, this study also calls for implementation of clinical trials assessing the use of intensified therapy for patients with DLBCL who fail to achieve an EMR or MMR and have a positive PET or CT scan.

1.
Schmitz R, Wright GW, Huang DW, et al.
Genetics and pathogenesis of diffuse large B-cell lymphoma.
N Engl J Med.
2018;378:1396-1407.
https://www.ncbi.nlm.nih.gov/pubmed/29641966
2.
Chapuy B, Stewart C, Dunford AJ, et al.
Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes.
Nat Med.
2018;24:679-690.
https://www.ncbi.nlm.nih.gov/pubmed/29713087
3.
Reddy A, Zhang J, Davis NS, et al.
Genetic and functional drivers of diffuse large B cell lymphoma.
Cell.
2017;171:481-494.e15.
https://www.ncbi.nlm.nih.gov/pubmed/28985567
4.
Newman AM, Bratman SV, To J, et al.
An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage.
Nat Med.
2014;20:548-554.
https://www.ncbi.nlm.nih.gov/pubmed/24705333

Competing Interests

Dr. Abdel-Wahab indicated no relevant conflicts of interest.