Response assessment in lymphoma relies on imaging scans that do not capture biologic processes at the molecular level. Monitoring circulating tumor DNA (ctDNA) with next-generation sequencing–based assays can detect recurrent disease prior to scans and “liquid biopsies” for somatic mutations address tumor heterogeneity, clonal evolution, and mechanisms of resistance to guide precision treatment. Preanalytic collection and processing procedures should be validated and standardized. We describe emerging applications of ctDNA monitoring including real-time analysis of tumor dynamics, preclinical disease detection, and precision-directed treatment paradigms.

Monitoring treatment response in non-Hodgkin lymphoma relies on computerized tomography (CT) and 18fluoro-2-deoxyglucose positron emission tomography (PET) scans. Imaging scans provide macro estimates of tumor volume and location, but do not significantly improve survival and are limited by cost and risk of ionizing radiation.1-3  Because disease recurrence originates from persistent tumor below limits of clinical detection, imaging scans are suboptimal for surveillance monitoring in curable lymphomas, such as diffuse large B-cell lymphoma (DLBCL).1  Lack of specificity limits the accuracy of PET scans for response-adapted approaches and surveillance, despite improved sensitivity over CT scans.4,5  Imaging scans are “snapshots” of clinically detectable tumor and cannot capture dynamic processes such as tumor response kinetics, clonal evolution, and cellular resistance.

With emergence of targeted therapy, clinically validated technology is needed to temporally assess the molecular heterogeneity across disease sites.6,7  Tissue biopsies determine molecular features of a tumor, but are prone to sampling bias and difficult to obtain serially. Circulating tumor DNA (ctDNA) can provide an average of the overall clonal heterogeneity and noninvasively assess molecular changes over time. Free from sampling bias of singular site biopsies, ctDNA integrates all genetic lesions and provides a more detailed view of the tumor.

Assessment of ctDNA is powerful technology that overcomes fundamental limitations of imaging scans and molecular analysis via tissue biopsies. In this review, we discuss the technical issues and present the promises of ctDNA for clinical application and novel research design.

Fragmented DNA from normal and diseased tissue is constantly shed into the bloodstream as cell-free DNA (cfDNA) through processes of apoptosis, necrosis, and secretion.8,9  Patients with cancer have higher overall levels of cfDNA than healthy people, but the fraction of DNA from malignant cells, captured as ctDNA, may be as low as 0.01%.10-12  Because ctDNA derives from tumor tissue, it is a highly specific tumor biomarker.13  It is present without detectable circulating tumor cells and correlates with tumor burden in early- and late-stage malignancies.14,15  Assays designed for molecular monitoring must accurately discriminate ctDNA from DNA that originates in nonmalignant tissue.16 

Molecular monitoring of lymphoma by ctDNA has gained momentum as a result of technical advances in detection capability and processing speeds (Table 1). Quantification of ctDNA in B-cell lymphomas using polymerase chain reaction (PCR) analysis of rearranged immunoglobulin heavy chains are subject to relatively low sensitivity and artifact, limiting their clinical utility.17  Modern platforms combine universal PCR primers for the VDJ regions of immunoglobulin receptors with next-generation sequencing (NGS), resulting in highly sensitive and specific detection of ctDNA in multiple B-cell lymphomas.15,18,19  As a surrogate for the entire tumor genome, ctDNA may also be analyzed for tumor-specific mutations, commonly referred to as a “liquid biopsy.”20,21  Modern NGS techniques have the necessary specificity, and bandwidth to identify genetic aberrations circulating at low allele frequencies.22,23 

Table 1

Methods for molecular monitoring of ctDNA for non-Hodgkin lymphoma

ctDNA of IgHctDNA of VDJ sequenceTumor genotypic ctDNA
Technique Allele-specific PCR PCR + NGS NGS 
Primary tumor required Yes No* No 
Sensitivity 1 in 105 1 in 106 Unknown 
Processing time Weeks 1-2 wk 1-2 wk 
Track clonal evolution No Limited Possible 
Track resistance No Limited Possible 
Potential Tumor-specific
Quantifiable 
Tumor-specific
Quantifiable
Universal primers
Rapid turnaround time 
Broad genomic coverage
Track clonal evolution
Track resistance mechanisms 
Limitations Not universal
Specific primers required
Limited foci assessed 
Limited genotypic information Low allele frequency
Molecular heterogeneity of tumor 
ctDNA of IgHctDNA of VDJ sequenceTumor genotypic ctDNA
Technique Allele-specific PCR PCR + NGS NGS 
Primary tumor required Yes No* No 
Sensitivity 1 in 105 1 in 106 Unknown 
Processing time Weeks 1-2 wk 1-2 wk 
Track clonal evolution No Limited Possible 
Track resistance No Limited Possible 
Potential Tumor-specific
Quantifiable 
Tumor-specific
Quantifiable
Universal primers
Rapid turnaround time 
Broad genomic coverage
Track clonal evolution
Track resistance mechanisms 
Limitations Not universal
Specific primers required
Limited foci assessed 
Limited genotypic information Low allele frequency
Molecular heterogeneity of tumor 

IgH, immunoglobulin heavy chain; NGS, next-generation sequencing; PCR, polymerase chain reaction; VDJ, variable-diversity-joining region of the immunoglobulin receptor.

*

Tumor clonotype can be determined without baseline tissue, but the yield is lower.

Mutational panels common to lymphoma subtypes would obviate the need for baseline tumor.

Sample size required to detect 1 cellular equivalent.

Methods to identify specific ctDNA sequences perform best on tumor biopsies. Clonal VDJ sequences can be determined from baseline tumor tissue in more than 85% of DLBCL cases, but with a significantly lower yield from blood samples.15,19,24  Targeted resequencing mutational panels for ctDNA are now emerging, but also require baseline biopsy samples to confirm tumor origin.22,23,25  Hence, quality DNA extraction from pretreatment tissue is important and is affected by low tumor content and necrosis.

Within peripheral blood samples, high rates of DNA fragmentation and low ratios of tumor DNA are methodologically challenging barriers. Normal DNA contamination from white blood cells is typically higher in the serum than plasma, and delay and temperature of blood samples before centrifugation affect DNA concentration.26  Specialized collection tubes (Streck) can reduce DNA degradation by nucleases and contamination from white blood cell DNA.27  Although successful analysis of ctDNA can be performed on stored serum samples, standardized collection procedures will optimize the analysis, particularly for detection of tumor mutations.26,28 

The complete molecular heterogeneity of a tumor cannot be adequately assessed by single or even multiple biopsies, whereas a “liquid biopsy” captures genetic information shed from all sites of disease.16  Recent reports demonstrate high concordance between mutations within tissue and targeted resequencing panels from ctDNA23,25  Analysis of ctDNA can also detect somatic mutations not identified in tumor biopsies. However, mutations with low allele frequencies are difficult to detect in the blood, suggesting that the different methods provide complementary information. Liquid biopsies of ctDNA can also be effective when needle biopsies are challenging, such as isolated central nervous system disease.15,19,29  Recent reports have demonstrated that tumor-associated mutations are detectable in the plasma and cerebral spinal fluid of patients with primary central nervous system lymphoma (including L.M.S. and W.H.W., unreported data).30 

In addition to spatial heterogeneity, lymphomas exhibit temporal heterogeneity and continuously evolve over time, particularly under treatment selection pressure (Figure 1). In the case of DLBCL, evidence suggests recurrent disease comprises multiple genetically distinct subclones. Serial analysis of ctDNA for mutation allele frequency can analyze clonal evolution in consecutive samples and reveal a shift in the dominant subclone with potential implications for treatment.31-33  Monitoring ctDNA is a promising method for assessing subclone allele frequency to overcome both spatial and temporal tumor heterogeneity.7 

Figure 1

Serial monitoring with circulating tumor DNA to guide precision medicine. Lymphomas are composed of multiple tumor clones and subclones that serially evolve over time, especially under the selective pressure of therapy. Liquid biopsies of ctDNA detect molecular features of resistant disease at the molecular level and can noninvasively genotype ctDNA throughout the disease course. Serial monitoring of ctDNA may be a powerful tool to address temporal heterogeneity of tumors, to detect clonal evolution, and to study mechanisms of treatment resistance. The timing and nature the dominant relapsing clone may guide precision treatment at relapse. As examples, patients who relapse with a myc rearrangement (green) might be offered targeted therapy with a BET inhibitor, whereas patients who relapse with a TP53 mutation (magenta) could be offered an MDM2 inhibitor.

Figure 1

Serial monitoring with circulating tumor DNA to guide precision medicine. Lymphomas are composed of multiple tumor clones and subclones that serially evolve over time, especially under the selective pressure of therapy. Liquid biopsies of ctDNA detect molecular features of resistant disease at the molecular level and can noninvasively genotype ctDNA throughout the disease course. Serial monitoring of ctDNA may be a powerful tool to address temporal heterogeneity of tumors, to detect clonal evolution, and to study mechanisms of treatment resistance. The timing and nature the dominant relapsing clone may guide precision treatment at relapse. As examples, patients who relapse with a myc rearrangement (green) might be offered targeted therapy with a BET inhibitor, whereas patients who relapse with a TP53 mutation (magenta) could be offered an MDM2 inhibitor.

Close modal

DLBCL is molecularly heterogeneous, but rearranged VDJ immunoglobulin receptor genes are unique to each patient’s tumor and are readily detectable by modern NGS platforms.34  Quantitative high-throughput methods that combine PCR-based amplification of immunoglobulin gene segments and NGS can detect and quantify ctDNA from B-cell lymphomas in the blood with a detection limit of 1 tumor cell equivalent per 106 diploid genomes.15,19 

A recent study in 126 patients with untreated DLBCL demonstrated that ctDNA of VDJ predicted early treatment failure and serial monitoring after therapy identified disease recurrence months before CT scans.15  Another study in a separate cohort of patients with DLBCL demonstrated that monitoring cell-free ctDNA of VDJ in plasma was more effective than monitoring circulating cells with the same assay.19  Notably, both studies showed a significant association between the quantity of pretreatment ctDNA and indices of tumor mass.

Clinical applications of ctDNA depend on the assay performed, the timing of the assay, and the goal of therapy (Table 2). The therapeutic goal for aggressive B-cell lymphomas, such as DLBCL, is cure. This requires eradication of all tumor clones, or disease recurrence is virtually inevitable. Molecular relapse of ctDNA heralds failure of curative treatment. The highly sensitive and quantitative characteristics of VDJ ctDNA lead to early detection of treatment failure compared with conventional imaging (Figure 2A).15,19  Indeed, identification of a molecular relapse allows institution of salvage treatment before the development of high disease burden that is required for clinical detection of recurrence.

Table 2

Clinical applications of circulating tumor DNA for non-Hodgkin lymphoma

PretreatmentReceiving treatmentEnd-of-treatmentAfter treatment
ctDNA of VDJ sequence Prognostic tool Quantitate tumor kinetics
Early treatment failure
Response-adapted therapy 
Molecular remission
Maintenance therapy decisions 
Early recurrence detection
Clonal evolution 
Tumor genotypic ctDNA Prognostic tool Quantitate tumor kinetics Molecular remission Early recurrence detection 
 Initial therapy choice Early treatment failure
Early resistance mechanisms
Response-adapted therapy 
Maintenance therapy decisions Clonal evolution
Late resistance mechanisms
Select next therapy 
PretreatmentReceiving treatmentEnd-of-treatmentAfter treatment
ctDNA of VDJ sequence Prognostic tool Quantitate tumor kinetics
Early treatment failure
Response-adapted therapy 
Molecular remission
Maintenance therapy decisions 
Early recurrence detection
Clonal evolution 
Tumor genotypic ctDNA Prognostic tool Quantitate tumor kinetics Molecular remission Early recurrence detection 
 Initial therapy choice Early treatment failure
Early resistance mechanisms
Response-adapted therapy 
Maintenance therapy decisions Clonal evolution
Late resistance mechanisms
Select next therapy 
Figure 2

Monitoring circulating tumor DNA enhances detection of relapse and defines molecular remission. The lead time offered by serial monitoring of ctDNA represents an opportunity for early intervention with minimal tumor burden. The clinical applications would differ when treating with curative intent (ie, aggressive lymphomas) or more extended duration of therapy (ie, indolent lymphomas). (A) Monitoring therapy for ctDNA for curative intent. The patient with no relapse (green) achieves a complete molecular remission and represents successful cure of lymphoma. The patient with late relapse (blue) initially achieves a complete molecular remission, but has ctDNA reappear before imaging, which creates a lead time for possible intervention. The patient with early relapse (red) has rising levels of ctDNA shortly after completion of therapy with a narrower lead time. The patient with primary refractory disease (brown) has persistence of minimal residual disease at the end of therapy that is undetectable by imaging. (B) Monitoring therapy with ctDNA for extended duration. Indolent lymphomas are frequently treated for extended durations with maintenance therapy designed to prolong duration of remission. Successful maintenance therapy (green) could be monitored with ctDNA and continued as long as disease remains undetectable. Patients who have the reappearance of ctDNA while receiving maintenance therapy (red) might be considered for alternative therapy before clinical effects. Patients who are not initially treated with maintenance therapy can be offered “delayed maintenance” at a time when disease is detectable by ctDNA, but not yet detectable by imaging scans.

Figure 2

Monitoring circulating tumor DNA enhances detection of relapse and defines molecular remission. The lead time offered by serial monitoring of ctDNA represents an opportunity for early intervention with minimal tumor burden. The clinical applications would differ when treating with curative intent (ie, aggressive lymphomas) or more extended duration of therapy (ie, indolent lymphomas). (A) Monitoring therapy for ctDNA for curative intent. The patient with no relapse (green) achieves a complete molecular remission and represents successful cure of lymphoma. The patient with late relapse (blue) initially achieves a complete molecular remission, but has ctDNA reappear before imaging, which creates a lead time for possible intervention. The patient with early relapse (red) has rising levels of ctDNA shortly after completion of therapy with a narrower lead time. The patient with primary refractory disease (brown) has persistence of minimal residual disease at the end of therapy that is undetectable by imaging. (B) Monitoring therapy with ctDNA for extended duration. Indolent lymphomas are frequently treated for extended durations with maintenance therapy designed to prolong duration of remission. Successful maintenance therapy (green) could be monitored with ctDNA and continued as long as disease remains undetectable. Patients who have the reappearance of ctDNA while receiving maintenance therapy (red) might be considered for alternative therapy before clinical effects. Patients who are not initially treated with maintenance therapy can be offered “delayed maintenance” at a time when disease is detectable by ctDNA, but not yet detectable by imaging scans.

Close modal

Indolent lymphomas present a different scenario because they generally are not treated with curative intent. In this setting, monitoring of ctDNA provides a quantitative estimate of sensitivity to treatment and can be monitored serially to guide treatment decisions. The common practice to treat indolent lymphomas for prolonged and even indefinite periods of time offers opportunities for ctDNA monitoring (Figure 2B). In some cases, extended treatment may be unnecessary, such as in patients who become ctDNA negative (ie, minimal residual disease negative), whereas in other cases, resistant clones may accumulate below the level of imaging detection. In both cases, serial monitoring of ctDNA could inform clinical decision-making, including treatment duration, retreatment timing, and guide precision treatment based on features of the dominant clones (Figure 1).

Pretreatment applications

ctDNA resequencing mutational panels (liquid biopsies) and quantification of pretreatment VDJ ctDNA provide novel opportunities for precision treatment. Multiple studies have shown a correlation between clinical determinants of tumor burden and quantitative ctDNA.14,15,19  A few studies have reported that higher pretreatment concentrations of ctDNA are associated with a poorer prognosis, possibly because of higher tumor burden, and hence the potential for more aggressive treatment.11,23  Molecular features such as tumor metabolism and/or proliferation may also be captured by ctDNA analysis and provide new areas of investigation. The molecular information obtained from liquid biopsies may provide valuable information on targetable oncogenic pathways and mutations, which are heterogeneous in DLBCL and often track with the cell of origin.6 

Response assessment

The current response criteria for lymphoma rely on CT and PET imaging, and the completeness of response to initial therapy is prognostic.35,36  Detection of subclinical disease by ctDNA, however, could identify patients not in complete molecular remission (Figure 2A). A recent study of ctDNA for tumor VDJ in DLBCL showed that patients who did not achieve a complete molecular remission were not cured, indicating its importance in aggressive lymphomas.15  For indolent lymphomas, achieving complete molecular remission may assist with treatment decisions regarding maintenance therapy and provide a novel endpoint for clinical trials and drug approval. In follicular lymphoma, PCR-based primers to detect BCL2 breakpoints have been used to assess molecular remission, which is associated with improved outcome.37,38  Similar findings have been reported in mantle cell lymphoma, using PCR assays to detect the CCND1/IgH translocations.39  These findings support the importance of molecular remissions.

Interim monitoring

Interim ctDNA monitoring (ie, during treatment), such as interim PET scans, can be developed for risk-adapted treatment strategies including early treatment termination for patients in molecular remission or treatment modification for patients with poorly responsive tumors. Although risk-adaptive strategies based on interim PET scans can identify patients with DLBCL at risk for treatment failure, 2 recent prospective studies failed to show clinical benefit from switching therapy, indicating the need for more sensitive and specific selection methods.40-42  Interim ctDNA monitoring has potential advantages over PET scans as a result of high tumor specificity, quantitative analysis, and temporal kinetics. Indeed, quantitative response kinetics may improve on response-adapted strategies in lymphomas. Our group studied the predictive significance of quantitative ctDNA for VDJ after each treatment cycle in previously untreated patients with DLBCL.15  Patients who cleared ctDNA after 2 cycles were significantly more likely to be progression-free at 5 years compared with patients who were ctDNA positive (80.2% vs 41.7%; P < .0001).15  Approximately half the patients cleared ctDNA after only 1 cycle, and 78% were negative after 2 treatment cycles. Various patterns of interim ctDNA kinetics were associated with early treatment failure, but absence of clearance was associated with the shortest survival.15  Other reports of interim ctDNA monitoring in DLBCL have also observed that ctDNA concentrations after only 1 to 2 cycles are predictive.22  Future studies are needed to validate the clinical use of interim ctDNA and its role with interim PET scans.

Posttreatment monitoring

The ability to identify patients with aggressive lymphomas before they develop clinically detectable disease has significant appeal because some may yet be cured with further treatment (Figure 2A). Although this has not been prospectively validated, it has been shown that patients with lower disease burdens at recurrence have better outcomes.43  The potential value of surveillance ctDNA monitoring lies in its low detection limit and ease of repetition. We showed that serial surveillance monitoring of ctDNA for VDJ after therapy in DLBCL detected recurrence a median of 3.5 month (range, 0-200 months) before CT scans.15  In patients who relapsed more than 6 months from the end of therapy, ctDNA was identified in 91% (10/11), and 80% (8/10) were detected before clinical disease on CT scans. In another series, similar rates of detection before relapse were observed.19  Paradigms for molecular surveillance monitoring will differ according to lymphoma subtype and therapeutic goals. For indolent lymphoma, the usual clinical goal is disease control, although the potential for cure cannot be ruled out. In the case of long-term disease control, despite the persistence of malignant clones, the goal of surveillance monitoring is to assess the malignant clone kinetics clonal evolution (Figure 1).

ctDNA and precision medicine

Identification of the molecular aberrations within an individual patient’s lymphoma will be needed for precision treatment decisions.6  Barriers to precision treatment include spatial and temporal tumor heterogeneity.7  Although in its infancy and unvalidated for clinical decision-making, the liquid biopsy has obvious advantages for selecting treatment based on the identification of dominant resistant clones (Figure 1). For example, increasing frequency of clones with myc translocations or TP53 mutations suggest emerging resistance, and may allow treatment modifications with targeted agents before the emergence of clinically detectable disease (Figure 1). Individual somatic mutations can also predict response to target agents, as recently shown with CD79B and MYD88 mutations in DLBCL and response to ibrutinib.44  Such information is likely to significantly improve outcomes through rational timing of treatment and precision drug selection. A new generation of “smart trials” based on this technology may also lead to more rational and rapid drug development and approval.

Circulating tumor DNA has the potential to transform clinical care paradigms and future trial designs. The ability of tumor VDJ ctDNA to dynamically assess molecular tumor response and detect recurrence of occult disease in B-cell lymphomas will undoubtedly improve the care of these patients. For indolent lymphomas, ctDNA provides a critical “look below the water line” that can aid clinical decisions on treatment timing and duration. Liquid biopsies of tumor genotypic ctDNA add a further dimension that can integrate all the genetic lesions within a tumor and optimally address traditional barriers to precision treatment such as tumor heterogeneity and clonal evolution. As we move forward, it will be imperative to standardize collection, storage, and processing procedures for ctDNA technology and validate its clinical and research utility.

We acknowledge support from the intramural research program of the National Institutes of Health, especially Kieron Dunleavy and the research team. Alan Hoofring and S. Peter Wu are acknowledged for illustration support.

Contribution: All authors (M.R., L.M.S., and W.H.W.) made a substantial contribution to discussion of the content. M.R. wrote the first draft of the manuscript, and W.H.W. revised the final draft. All authors reviewed and edited the final manuscript before submission.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Mark Roschewski, Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bldg 10, Room 4N/115, Bethesda, MD 20892; e-mail mark.roschewski@nih.gov.

1
Thompson
 
CA
Ghesquieres
 
H
Maurer
 
MJ
, et al. 
Utility of routine post-therapy surveillance imaging in diffuse large B-cell lymphoma.
J Clin Oncol
2014
, vol. 
32
 
31
(pg. 
3506
-
3512
)
2
Huntington
 
SF
Svoboda
 
J
Doshi
 
JA
Cost-effectiveness analysis of routine surveillance imaging of patients with diffuse large B-cell lymphoma in first remission.
J Clin Oncol
2015
, vol. 
33
 
13
(pg. 
1467
-
1474
)
3
Brenner
 
DJ
Hall
 
EJ
Computed tomography--an increasing source of radiation exposure.
N Engl J Med
2007
, vol. 
357
 
22
(pg. 
2277
-
2284
)
4
Moskowitz
 
CH
Schöder
 
H
Teruya-Feldstein
 
J
, et al. 
Risk-adapted dose-dense immunochemotherapy determined by interim FDG-PET in advanced-stage diffuse large B-cell lymphoma.
J Clin Oncol
2010
, vol. 
28
 
11
(pg. 
1896
-
1903
)
5
Cheah
 
CY
Hofman
 
MS
Dickinson
 
M
, et al. 
Limited role for surveillance PET-CT scanning in patients with diffuse large B-cell lymphoma in complete metabolic remission following primary therapy.
Br J Cancer
2013
, vol. 
109
 
2
(pg. 
312
-
317
)
6
Roschewski
 
M
Staudt
 
LM
Wilson
 
WH
Diffuse large B-cell lymphoma-treatment approaches in the molecular era.
Nat Rev Clin Oncol
2014
, vol. 
11
 
1
(pg. 
12
-
23
)
7
Alizadeh
 
AA
Aranda
 
V
Bardelli
 
A
, et al. 
Toward understanding and exploiting tumor heterogeneity.
Nat Med
2015
, vol. 
21
 
8
(pg. 
846
-
853
)
8
Mandel
 
P
Metais
 
P
Les acides nucléiques du plasma sanguin chez l'homme [in French].
C R Seances Soc Biol Fil
1948
, vol. 
142
 
3-4
(pg. 
241
-
243
)
9
Jahr
 
S
Hentze
 
H
Englisch
 
S
, et al. 
DNA fragments in the blood plasma of cancer patients: quantitations and evidence for their origin from apoptotic and necrotic cells.
Cancer Res
2001
, vol. 
61
 
4
(pg. 
1659
-
1665
)
10
Leon
 
SA
Shapiro
 
B
Sklaroff
 
DM
Yaros
 
MJ
Free DNA in the serum of cancer patients and the effect of therapy.
Cancer Res
1977
, vol. 
37
 
3
(pg. 
646
-
650
)
11
Hohaus
 
S
Giachelia
 
M
Massini
 
G
, et al. 
Cell-free circulating DNA in Hodgkin’s and non-Hodgkin’s lymphomas.
Ann Oncol
2009
, vol. 
20
 
8
(pg. 
1408
-
1413
)
12
Schwarzenbach
 
H
Hoon
 
DS
Pantel
 
K
Cell-free nucleic acids as biomarkers in cancer patients.
Nat Rev Cancer
2011
, vol. 
11
 
6
(pg. 
426
-
437
)
13
Bohers
 
E
Viailly
 
PJ
Dubois
 
S
, et al. 
Somatic mutations of cell-free circulating DNA detected by next-generation sequencing reflect the genetic changes in both germinal center B-cell-like and activated B-cell-like diffuse large B-cell lymphomas at the time of diagnosis.
Haematologica
2015
, vol. 
100
 
7
(pg. 
e280
-
e284
)
14
Bettegowda
 
C
Sausen
 
M
Leary
 
RJ
, et al. 
Detection of circulating tumor DNA in early- and late-stage human malignancies.
Sci Transl Med
2014
, vol. 
6
 
224
pg. 
224ra24
 
15
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
, vol. 
16
 
5
(pg. 
541
-
549
)
16
Diaz
 
LA
Bardelli
 
A
Liquid biopsies: genotyping circulating tumor DNA.
J Clin Oncol
2014
, vol. 
32
 
6
(pg. 
579
-
586
)
17
Frickhofen
 
N
Müller
 
E
Sandherr
 
M
, et al. 
Rearranged Ig heavy chain DNA is detectable in cell-free blood samples of patients with B-cell neoplasia.
Blood
1997
, vol. 
90
 
12
(pg. 
4953
-
4960
)
18
Ladetto
 
M
Brüggemann
 
M
Monitillo
 
L
, et al. 
Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders.
Leukemia
2014
, vol. 
28
 
6
(pg. 
1299
-
1307
)
19
Kurtz
 
DM
Green
 
MR
Bratman
 
SV
, et al. 
Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing.
Blood
2015
, vol. 
125
 
24
(pg. 
3679
-
3687
)
20
Forshew
 
T
Murtaza
 
M
Parkinson
 
C
, et al. 
Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA.
Sci Transl Med
2012
, vol. 
4
 
136
pg. 
136ra68
 
21
Newman
 
AM
Bratman
 
SV
To
 
J
, et al. 
An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage.
Nat Med
2014
, vol. 
20
 
5
(pg. 
548
-
554
)
22
Kurtz
 
DM
Scherer
 
F
Newman
 
AM
, et al. 
Dynamic noninvasive genomic monitoring for outcome prediction in diffuse large B-cell lymphoma [abstract].
Blood
2015
, vol. 
126
 
23
 
Abstract 130
23
Scherer
 
F
Kurtz
 
DM
Newman
 
AM
, et al. 
Noninvasive genotyping and assessment of treatment response in diffuse large B cell lymphoma [abstract].
Blood
2015
, vol. 
126
 
23
 
Abstract 114
24
Armand
 
P
Oki
 
Y
Neuberg
 
DS
, et al. 
Detection of circulating tumour DNA in patients with aggressive B-cell non-Hodgkin lymphoma.
Br J Haematol
2013
, vol. 
163
 
1
(pg. 
123
-
126
)
25
Rasi
 
S
Monti
 
S
Zanni
 
M
, et al. 
Liquid biopsy as a tool for monitoring the genotype of diffuse large B-cell lymphoma [abstract].
Blood
2015
, vol. 
126
 
23
 
Abstract 127
26
El Messaoudi
 
S
Rolet
 
F
Mouliere
 
F
Thierry
 
AR
Circulating cell free DNA: Preanalytical considerations.
Clin Chim Acta
2013
, vol. 
424
 (pg. 
222
-
230
)
27
Qin
 
J
Williams
 
TL
Fernando
 
MR
A novel blood collection device stabilizes cell-free RNA in blood during sample shipping and storage.
BMC Res Notes
2013
, vol. 
6
 pg. 
380
 
28
Heitzer
 
E
Ulz
 
P
Geigl
 
JB
Circulating tumor DNA as a liquid biopsy for cancer.
Clin Chem
2015
, vol. 
61
 
1
(pg. 
112
-
123
)
29
Wang
 
Y
Springer
 
S
Zhang
 
M
, et al. 
Detection of tumor-derived DNA in cerebrospinal fluid of patients with primary tumors of the brain and spinal cord.
Proc Natl Acad Sci USA
2015
, vol. 
112
 
31
(pg. 
9704
-
9709
)
30
Fontanilles
 
M
Marguet
 
F
Bohers
 
É
, et al. 
Somatic mutations detected in plasma cell-free DNA by targeted sequencing: assessment of liquid biopsy in primary central nervous system lymphoma [abstract].
Blood
2015
, vol. 
126
 
23
 
Abstract 332
31
Meador
 
CB
Lovly
 
CM
Liquid biopsies reveal the dynamic nature of resistance mechanisms in solid tumors.
Nat Med
2015
, vol. 
21
 
7
(pg. 
663
-
665
)
32
Frenel
 
JS
Carreira
 
S
Goodall
 
J
, et al. 
Serial Next-Generation Sequencing of Circulating Cell-Free DNA Evaluating Tumor Clone Response To Molecularly Targeted Drug Administration.
Clin Cancer Res
2015
, vol. 
21
 
20
(pg. 
4586
-
4596
)
33
Murtaza
 
M
Dawson
 
SJ
Tsui
 
DW
, et al. 
Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA.
Nature
2013
, vol. 
497
 
7447
(pg. 
108
-
112
)
34
Arnold
 
A
Cossman
 
J
Bakhshi
 
A
Jaffe
 
ES
Waldmann
 
TA
Korsmeyer
 
SJ
Immunoglobulin-gene rearrangements as unique clonal markers in human lymphoid neoplasms.
N Engl J Med
1983
, vol. 
309
 
26
(pg. 
1593
-
1599
)
35
Adams
 
HJ
Nievelstein
 
RA
Kwee
 
TC
Prognostic value of complete remission status at end-of-treatment FDG-PET in R-CHOP-treated diffuse large B-cell lymphoma: systematic review and meta-analysis.
Br J Haematol
2015
, vol. 
170
 
2
(pg. 
185
-
191
)
36
Trotman
 
J
Luminari
 
S
Boussetta
 
S
, et al. 
Prognostic value of PET-CT after first-line therapy in patients with follicular lymphoma: a pooled analysis of central scan review in three multicentre studies.
Lancet Haematol
2014
, vol. 
1
 
1
(pg. 
e17
-
e27
)
37
Ladetto
 
M
Lobetti-Bodoni
 
C
Mantoan
 
B
, et al. 
Fondazione Italiana Linfomi
Persistence of minimal residual disease in bone marrow predicts outcome in follicular lymphomas treated with a rituximab-intensive program.
Blood
2013
, vol. 
122
 
23
(pg. 
3759
-
3766
)
38
Galimberti
 
S
Luminari
 
S
Ciabatti
 
E
, et al. 
Minimal residual disease after conventional treatment significantly impacts on progression-free survival of patients with follicular lymphoma: the FIL FOLL05 trial.
Clin Cancer Res
2014
, vol. 
20
 
24
(pg. 
6398
-
6405
)
39
Pott
 
C
Hoster
 
E
Delfau-Larue
 
MH
, et al. 
Molecular remission is an independent predictor of clinical outcome in patients with mantle cell lymphoma after combined immunochemotherapy: a European MCL intergroup study.
Blood
2010
, vol. 
115
 
16
(pg. 
3215
-
3223
)
40
Duehrsen
 
U
Hüttmann
 
A
Müller
 
S
, et al. 
Positron emission tomography (PET) guided therapy of aggressive lymphomas—a randomized controlled trial comparing different treatment approaches based on interim PET results (PETAL trial) [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 391
41
Sehn
 
LH
Hardy
 
ELG
Gill
 
KK
, et al. 
Phase 2 trial of interim PET scan-tailored therapy in patients with advanced stage diffuse large B-cell lymphoma (DLBCL) in British Columbia (BC) [abstract].
Blood
2014
, vol. 
124
 
21
 
Abstract 392
42
Mamot
 
C
Klingbiel
 
D
Hitz
 
F
, et al. 
Final Results of a Prospective Evaluation of the Predictive Value of Interim Positron Emission Tomography in Patients With Diffuse Large B-Cell Lymphoma Treated With R-CHOP-14 (SAKK 38/07).
J Clin Oncol
2015
, vol. 
33
 
23
(pg. 
2523
-
2529
)
43
Hamlin
 
PA
Zelenetz
 
AD
Kewalramani
 
T
, et al. 
Age-adjusted International Prognostic Index predicts autologous stem cell transplantation outcome for patients with relapsed or primary refractory diffuse large B-cell lymphoma.
Blood
2003
, vol. 
102
 
6
(pg. 
1989
-
1996
)
44
Wilson
 
WH
Young
 
RM
Schmitz
 
R
, et al. 
Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma.
Nat Med
2015
, vol. 
21
 
8
(pg. 
922
-
926
)
Sign in via your Institution