• Non ci sono risultati.

Highly sensitive MYD88 l265p mutation detection by droplet digital polymerase chain reaction in waldenström macroglobulinemia

N/A
N/A
Protected

Academic year: 2021

Condividi "Highly sensitive MYD88 l265p mutation detection by droplet digital polymerase chain reaction in waldenström macroglobulinemia"

Copied!
50
0
0

Testo completo

(1)

07 August 2021

AperTO - Archivio Istituzionale Open Access dell'Università di Torino

Original Citation:

Highly sensitive MYD88 l265p mutation detection by droplet digital polymerase chain reaction in waldenström macroglobulinemia

Published version:

DOI:10.3324/haematol.2017.186528 Terms of use:

Open Access

(Article begins on next page)

Anyone can freely access the full text of works made available as "Open Access". Works made available under a Creative Commons license can be used according to the terms and conditions of said license. Use of all other works requires consent of the right holder (author or publisher) if not exempted from copyright protection by the applicable law. Availability:

This is the author's manuscript

(2)

Highly sensitive MYD88

L265P

mutation detection by droplet digital

PCR in Waldenström Macroglobulinemia

by Daniela Drandi, Elisa Genuardi, Irene Dogliotti, Martina Ferrante, Cristina Jiménez, Francesca Guerrini, Mariella Lo Schirico, Barbara Mantoan, Vittorio Muccio, Giuseppe Lia, Gian Maria Zaccaria, Paola Omedè, Roberto Passera, Lorella Orsucci, Giulia Benevolo, Federica Cavallo, Sara Galimberti, Ramón García-Sanz, Mario Boccadoro, Marco Ladetto, and Simone Ferrero

Haematologica 2018 [Epub ahead of print]

Citation: Daniela Drandi, Elisa Genuardi, Irene Dogliotti, Martina Ferrante, Cristina Jiménez, Francesca Guerrini, Mariella Lo Schirico, Barbara Mantoan, Vittorio Muccio, Giuseppe Lia,

Gian Maria Zaccaria, Paola Omedè, Roberto Passera, Lorella Orsucci, Giulia Benevolo, Federica Cavallo, Sara Galimberti, Ramón García-Sanz, Mario Boccadoro, Marco Ladetto, and Simone Ferrero.

Highly sensitive MYD88L265P mutation detection by droplet digital PCR in Waldenström Macroglobulinemia. Haematologica. 2018; 103:xxx

doi:10.3324/haematol.2017.186528 Publisher's Disclaimer.

E-publishing ahead of print is increasingly important for the rapid dissemination of science. Haematologica is, therefore, E-publishing PDF files of an early version of manuscripts that have completed a regular peer review and have been accepted for publication. E-publishing of this PDF file has been approved by the authors. After having E-published Ahead of Print, manuscripts will then undergo technical and English editing, typesetting, proof correction and be presented for the authors' final approval; the final version of the manuscript will then appear in print on a regular issue of the journal. All legal disclaimers that apply to the journal also pertain to this production process.

Copyright 2018 Ferrata Storti Foundation.

(3)

1

Highly sensitive MYD88

L265P

mutation detection by droplet digital PCR in

Waldenström Macroglobulinemia

Daniela Drandi1, Elisa Genuardi1, Irene Dogliotti1,2, Martina Ferrante1, Cristina Jiménez3, Francesca Guerrini4, Mariella Lo Schirico1, Barbara Mantoan1, Vittorio Muccio2, Giuseppe Lia1, Gian Maria Zaccaria1,5, Paola Omedè2, Roberto Passera6, Lorella Orsucci7, Giulia Benevolo7, Federica Cavallo1,2, Sara Galimberti4, Ramón García Sanz3, Mario Boccadoro1,2, Marco Ladetto8 and Simone Ferrero1,2

1

Department of Molecular Biotechnologies and Health Sciences, Hematology Division, University of Torino, Torino, Italy;

2

Division of Hematology 1U, AOU “Città della Salute e della Scienza di Torino”, Torino, Italy;

3

Hematology Department, University Hospital of Salamanca and Research Biomedical Institute of Salamanca (IBSAL), Salamanca, Spain;

4

Division of Hematology, Department of Oncology, Santa Chiara Hospital, Pisa, Italy.

5

Biolab, Department of Electronics and Telecommunications, Politecnico di Torino, Torino, Italy;

6

Biostatistics unit, Division of Nuclear Medicine, AOU Città della Salute e della Scienza di Torino, Torino, Italy

7

Division of Hematology 2, AOU “Città della Salute e della Scienza di Torino”, Torino, Italy;

8

Division of Hematology, AO SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy

Runninig title: MYD88L265P mutation detection by ddPCR on BM, PB and ctDNA

Corresponding author: Daniela Drandi - Department of Molecular Biotechnology and

Health sciences, Hematology Division, via Genova 3, 10126 Torino, Italy, Phone: +39 011 6336884; Fax +39 011 6963737 E-mail: daniela.drandi@unito.it

Word count:

Abstract: 246- Introduction: 391- Methods: 481- Results:1562- Discussion: 1158

Total number of figures and tables: 5 figures and 1 table (4 supplementary tables and 7

(4)

2

Abstract

We here describe a novel method for MYD88L265P mutation detection and minimal residual disease monitoring in Waldenström Macroglobulinemia, by droplet digital PCR, in bone marrow and peripheral blood cells, as well as in circulating cell-free DNA. Our method shows a sensitivity of 5.00E−05, by far superior to the widely used allele-specific polymerase chain reaction (1.00E-03). Overall, 291 unsorted samples from 148 patients (133 Waldenström, 11 IgG-lymphoplasmacytic lymphoma and 4 IgM-monoclonal gammopathy of undetermined significance), 194 baseline and 97 follow-up, were analyzed. 122/128 (95.3%) bone marrow and 47/66 (71.2%) baseline peripheral blood samples scored positive for MYD88L265P. Moreover, to investigate whether MYD88L265P by droplet digital PCR could be used for minimal residual disease monitoring, mutation levels were compared with IGH-based minimal residual disease analysis in 10 patients, showing to be as informative as to the classical, standardized but not yet validated in Waldenström Macroglobulinemia, IGH-based minimal residual disease assay (r2=0.64). Finally, MYD88L265P detection performed by droplet digital PCR on plasmatic circulating tumor DNA from 60 patients showed a good correlation with bone marrow (bone marrow median mutational value 1.92E-02, plasmatic circulating tumor DNA value: 1.4E-02, peripheral blood value: 1.03E-03). This study indicates that droplet digital PCR MYD88L265P assay is a feasible and sensitive tool for mutational screening and minimal residual disease monitoring in Waldenström Macroglobulinemia. Both unsorted bone marrow and peripheral blood samples can be reliably tested, as well as circulating tumor DNA, that represents an attractive, less invasive alternative to bone marrow for MYD88L265P detection.

(5)

3

INTRODUCTION

In the latest years, MYD88L265P mutation has been recurrently identified in Waldenström macroglobulinemia (WM)1,2, an indolent lymphoplasmacytic lymphoma characterized by the accumulation in the bone marrow (BM) of monoclonal lymphocytes, lymphoplasmacytic cells and plasma cells, responsible for monoclonal IgM protein secretion.3 Several studies using different techniques such as Sanger sequencing, polymerase chain reaction (PCR) and allele-specific quantitative PCR (ASqPCR), on CD19-sorted BM samples, observed that about 90% of WM patients carry the MYD88L265P mutation, while it is present in 14-29% of “activated B-cell” diffuse large B-cell lymphomas (ABC-DLBCL),4,5 6-10% of marginal zone lymphoma (MZL), 3-5% of chronic lymphocytic leukemia (CLL) and it is absent in multiple myeloma (MM) and in non-IgM monoclonal gammopathies of uncertain significance (MGUS).6–8 Therefore, MYD88L265P is now considered a hallmark of WM and it might be helpful in the differential diagnosis with other lymphoproliferative neoplasms with overlapping clinical features, such as MM.9,10 Furthermore, it might represent an ideal marker for minimal residual disease (MRD) monitoring in a disease whose therapeutic scenario is rapidly changing, with many new available and highly effective drugs.11–17 Moreover, MYD88L265P has been demonstrated on CD19-sorted BM samples in 50-80% of patients with IgM-MGUS, an asymptomatic phase which represents a pre-neoplastic condition, postulating a potential role in disease progression.6,8,18–20 BM biopsy is mandatory for differential diagnosis between WM and IgM-MGUS, but patients with asymptomatic M component do not easily accept to undergo such an invasive procedure. The availability of accurate diagnostic tools on peripheral blood (PB), or even on urine samples, could overcome this problem and avoid the risk of patients misclassification. Additionally, the current MYD88L265P ASqPCR method, lacks sensitivity (1.00E-03) and thus

(6)

4

is not suitable for MRD.6,21 Indeed, ASqPCR is suboptimal for testing specimens like unsorted BM or even PB, which harbors low concentrations of circulating tumor cells (especially after immunochemotherapy), neither to assess cell-free tumor DNA (ctDNA), usually present at very low amount in plasma, including liquor and pleural effusion.22,23 Recently, digital PCR has been shown to be a powerful technique that provides improved sensitivity, precision and reproducibility overcoming some of qPCR pitfalls.24,25 We here describe a newly developed, highly sensitive, droplet digital PCR (ddPCR) assay for the identification of the MYD88L265P mutation in patients affected by WM or LPL, suitable for screening and MRD monitoring on BM and PB cells, as well as on cell-free DNA.

METHODS

Patients and samples collection

BM, PB, plasma and urine samples were collected at baseline and during follow-up (FU) from patients affected by WM, IgM-MGUS and IgG secreting LPL (Figure S1). Patients have been consecutively included in this study and were classified based on the 2008 WHO classification criteria (3). PB from 40 healthy subjects and BM from 20 MM patients were used as negative controls. Samples collection and storage as well as nucleic acid extraction procedures are described in the Online Supplement.

All patients provided written informed consent for the research use of their biological samples, in accordance with Institutional Review Boards and the Helsinki's declaration. This study was approved by the local Ethics Committee.

(7)

5

ddPCR assays for MYD88L265P mutation detection

Mutation detection assay was designed as reported in Figure S2A. ddPCR was performed using the QX100 Droplet Digital PCR system (Bio-Rad Laboratories) as detailed reported in the Online Supplement. Samples were tested in triplicate and results expressed as merge of wells. The cut-off for mutation was set based on the highest MYD88L265P level detected within the control group and is indicated in figures as a dashed line. Samples with ratio value below the dashed line are considered MYD88L265P WT. Each experiment included a no template control (NTC), a known highly mutated positive control sample (mutation rate = 70%, MUT/WT ratio 7x10-1), previously tested by Sanger sequencing, as reported by Treon et al1, and a negative control (healthy donor or MM gDNA). dMIQE guidelines (Minimum Information for Publication of Quantitative Digital PCR Experiments) for ddPCR experiments are listed in Table S1.25

ASqPCR assay for MYD88L265P mutation detection

The sensitivity of MYD88L265P ddPCR was compared to the ASqPCR assay previously described by Xu et al.6 on a ten-fold serial dilutions standard curve. Moreover, 100 WM samples from the Torino series and 23 patients from the Salamanca series were analyzed in parallel, following the strategy described by Jiménez et al.21 Additionally, MYD88L265P mutation by ddPCR was compared to qBiomarker™ Somatic Mutation Assay kit for MYD88_85940 (Qiagen) on 15 samples from University of Pisa.

ASOqPCR assay for tumor-specific IGH-VDJ rearrangement

In order to perform a comparison to the worldwide standardized ASOqPCR technique for immunoglobulin-VDJ rearrangement (IGH-VDJ) MRD analysis, patient-specific IGH-VDJ

(8)

6

were amplified and directly sequenced26 (Figure S2B). IGH-based MRD analysis was performed according to the Euro-MRD guidelines.27

Statistical analyses

The associations between categorical variables were analyzed by the Fisher’ exact test, while the Mann-Whitney and Kruskal-Wallis ones were used for the inference on continuous variables. All continuous variable results were expressed as the median (range); ddPCR and ASqPCR results were expressed as MUT/WT ratio. The interrater agreement on categorical data was estimated by computing the Fleiss' Kappa index. All reported p-values were estimated by the 2-sided exact method at the conventional 5% significance level. Data were analyzed as of July 2017 by R 3.4.1 (R Foundation for Statistical Computing, Vienna, Austria).28

RESULTS

Patients and samples

Overall, 291 samples of 148 patients from the three series (133 WM, 1 amyloid-associated IgM-LPL, 10 IgG-LPL, 4 IgM-MGUS) were analyzed: 194 samples were taken at baseline in active disease (128 BM, 66 PB) and 97 samples during follow-up (FU, 43 BM and 54 PB). The baseline samples were defined either “treatment-naïve” (TN) (143/194, 73.7%, 99 BM and 44 PB) or “relapsed” (RE) (51/194, 26.2%, 29 BM and 22 PB), according to previous exposure to chemo-immunotherapy. On the other hand, all the FU samples were taken for MRD purposes at different time points after specific treatment. An overview of the study

(9)

7

population distribution is summarized in Figure S1, while patients’ main clinical features are detailed in Table 1.

Detection limit of ddPCR

The detection limit (LOD) of ddPCR was determined using a serial dilution of MYD88L265P mutated gDNA in a WT DNA at level of 35, 3.5, 0.35, 0.035 and 0.0035 % corresponding to 10500, 1050, 105, 10.5 and 1 mutated copy numbers presented in 100 ng of gDNA (30000 copies) which is the overall quantity of gDNA loaded per well. We identified the LOD, using a statistical method based on binomial distribution, as previously reported29. This analysis indicated that we were able to detect the mutation, with a good grade of confidence, for more than 0,035% mutated copies (10 MUT copies in 30000 WT). This value mirrored the cut-off MUT/WT ratio we identified based on the control group (40 healthy subjects) (Figure 1A). Additional reproducibility tests confirmed that the above calculated LOD and the experimentally detected cut-off, arising from the control group, are equally reliable (see supplemental Table S2).

MYD88L265P screening by ddPCR in baseline samples

Overall 142/148 (96%) patients were identified as MYD88L265P mutated. We observed a 91.6% mutation rate (33/36) among RE and 97.3% rate (109/112) among TN patients (p=0.15). Notably, all the 6 MYD88L265P WT patients (4 WM with histological BM invasion of 20%, 30%, 30% and 60%) and 2 LPL IgG (with 20% and 30% histological BM invasion), showed neither alternative MYD88 nor CXCR4 mutations, as investigated by Sanger sequencing on unselected cells.10,12,30 All BM samples from 20 MM patients used as negative controls, were below the LOD (defined as previously described) and below the

(10)

8

MUT cut-off ratio setted based on 40 PB samples from healthy individuals (MUT/WT ratio <3.4E-04) (Figure 1A). Moreover, to confirm the specificity of our assay for mutational

screening, 15 mantle cell lymphoma (MCL), 10 follicular lymphoma (FL) and 10 CLL patients were tested for MYD88L265P mutation. All FL and MCL samples were WT, while 1 out of 10 CLL patients showed a mutation ratio of 4.4E-04, as already described in this disease.6

Looking at the single tissues, 95.3% (122/128) of baseline BM and 71.2% (47/66) of PB samples scored positive for MYD88L265P (BM median mutation burden 3.60E-02, range: 2.00E-04 – 7.30E-01; PB median 5.00E-03, range: 1.00E-04 – 2.80E-01) (Figure 1A). Notably, PB samples showed a statistically significant difference in mutation rate between RE (median burden 4.00E-04, range: 1.00E-04 – 1.00E-03) and TN (median burden 2.80E-03, range: 2.00E-04 – 1.00E-02) groups, supporting the possible negative impact, on mutation detection, of previous treatment on PB samples (p<0.0001) (Figure 1B).

Among this series, 74 patients presented BM/PB paired baseline samples. Overall, in this group of patients, the rate of MYD88L265P mutation detection by ddPCR is 93% (69/74) on BM and 72% (53/74) on PB samples. Accordingly, the detection rates are higher among TN than RE patients: 95% (52/55) on BM and 82% (45/55) on PB vs 89% (17/19) on BM and 42% (8/19) on PB, respectively (p=0.014). In addition, 15/74 patients (20%) showed a discordant BM-MUT/PB-WT match, being these discordances more frequent among RE (8/19, 42%) than TN cases (7/55 13%), p=0.02 (Figure 1C).

Finally, a reproducibility test within runs showed uniform results across the experiments, with inter-assay standard deviation (SD) of 1.3% for the MYD88L265P positive control and 0.01% for WT gDNA samples.

(11)

9

Comparison of MYD88L265P ddPCR versus ASqPCR assays

Once the ddPCR assay was optimized, the sensitivity of MYD88L265P ddPCR was compared to ASqPCR on a ten-fold serial dilutions standard curve built with highly MYD88L265P mutated WM sample, (70%, MUT/WT ratio 7.00E-01, diluted to 35%), previously identified by Sanger sequencing.1 Whereas ASqPCR standard curve confirmed the reported sensitivity of 1.00E−03 (0.25% out of 6000 WT),6 ddPCR reached a 1.5 log higher sensitivity (0.0035% out of 30000 WT) also due to higher amount of gDNA used in ddPCR compared to ASqPCR (100 ng vs 20ng) (Figure 2, Table S2). Overall 100 WM samples (48 BM, 52 PB, 40 baseline and 60 FU) from 48 patients, as well 20 control samples (15 healthy subjects and 5 MM) were tested by both methods (Figure S3). An example of mutation analysis performed by both methods, on two patients, is described in Figure S4. 35/40 (87.5%) baseline samples scored positive and 4/40 (10%) negative for MYD88L265P by both ddPCR and ASqPCR (being only one discordant ddPCR+/ASqPCR- case), while higher number of discordances were observed among FU samples: 13/60 (21.7%) ddPCR+/ASqPCR-, and 11/60 (18.3%) ddPCR-/ASqPCR+ (Table S3). Indeed, the strength of agreement resulted to be very good in the baseline cases (Cohen's K=0.8) while poor (Cohen's K=0.2) in FU samples, resembling what previously shown for low burden infiltrated samples.31 All control samples scored negative by both methods (median: ddPCR 1.75E-04 (range 3.10E-04 – 2.70E-05) and ASqPCR ΔCT=10 (range ΔCT=8.4-10.7, setting ΔCT=8 as cut-off for negativity).

Comparison of MYD88L265P and IGH-based ddPCR assays for MRD purposes

To investigate whether the MYD88L265P by ddPCR could be used for MRD detection, we compared it to the highly sensitive IGH-based MRD ddPCR assay. IGH rearrangement screening was performed only for patients with available FU samples. A clonal VDJ

(12)

10

rearrangement was detected in 34/52 (65%) patients, as expected in WM.32 All these patients scored positive for MYD88L265P by ddPCR. Therefore, we tested 10 informative patients, at baseline and during clinical FU, with both techniques. Overall, the 23 samples (18 BM, 5 PB) showed a good concordance (r2= 0.64) between the two methods (p<0.0001) (Figure 3).

MYD88L265P ddPCR on ctDNA

In order to investigate the feasibility and the sensitivity advantages of ddPCR based MYD88L265P mutation detection on plasma (PL) ctDNA versus PB gDNA, paired samples from 60 patients were analyzed. Interestingly, higher median levels of MYD88L265P MUT/WT ratio were detected in PL ctDNA (1.4E-02), if compared to PB (1.03E-03) (p<0.001) (Figure S5), while no statistically significant difference was observed between ctDNA and BM samples from 32 patients (1.92E-02 vs 1.4E-02) (p=0.2). Figure 4 shows the matches among BM-PL-PB MYD88L265P mutation quantification. Of note, 2 samples scored WT in all 3 compartments (Figure 4, Figure S6), while in 3 other patients MYD88L265P was detected in BM and PB, but scored WT in PL ctDNA: these samples have been excluded from the analysis since negativity, confirmed by RNAseP analysis, was ascribed to pre-analytical issues, related to sample collection and storage. Two cases were erroneously collected in lithium-heparin tubes instead of K3EDTA, while one case was properly collected but

processed more than 6 hours from blood draw, confirming the importance of collection tubes and timing processing for a good quality ctDNA, as already reported.33–35

(13)

11

To explore the potential role of MYD88L265P mutation detection by ddPCR for MRD monitoring and therapy response evaluation, we focused our analysis on 52 patients, which had at least one available FU sample. Patients features and treatment received are described in Table S4. The MRD monitoring of this series is graphically depicted in Figure 5. All the TN patients were MYD88L265P mutated at baseline (either on BM or PB or both), while two RE patients (one BM and one PB) scored WT (as false negative results, due to previous rituximab exposure, see below). After therapy, 6/22 informative cases showed a MRD negativization on BM (27%) and 17/35 (49%) on PB, further suggesting a faster MRD clearance in PB than in BM. Moreover, in the subsequent time points, the majority of patients remain MRD positive (namely, patients 26, 28-31 and 33-35), some remain MRD negative (patients 14-16, 37, 46 and 48), while a few show alternating results (patients 32, 40 and 50). Due to the limited and retrospective nature of the patients series, it is difficult to draw conclusions on the efficacy of single therapeutic schedules in clearing MRD in WM. However, the high potential of MRD shrinkage of regimens containing either fludarabine, bendamustine (each combined with rituximab) is highlighted in the Supplemental Material and Methods (Figures S7A-C).

Of note, two false negative cases can be highlighted in this series among RE patients. Namely, patient 44 showed a baseline BM sample depicted as WT (that means “MRD negative”), but with a borderline MUT/WT ratio, followed by a PB positivity at first FU (taken after three years). Moreover, another false negativity was observed on patient 45 PB baseline RE sample, reverting to positive during the subsequent FU (“MRD reappearance”).

Additionally, 27 paired PB/PL FU samples highlight the role that ctDNA might have for MYD88L265P mutation detection in pre-treated cases, as well as for MRD monitoring. Of note, 14/27 were concordantly positive and 3/27 concordantly negative, while 9/27 scored

(14)

12

positive only in PL and 1/27 positive on PB but not on PL. Interestingly, the 9 PL positive cases “rescued” by ctDNA analysis showed MYD88L265P levels in PL comparable to those detected in paired BM samples (see Figure 5: patients 52, 50 and 37).

DISCUSSION

ASqPCR has been widely used for MYD88L265P detection in WM, granting a higher level of sensitivity over Sanger sequencing.6,21 However, the recently developed ddPCR allowed to simplify and to improve the accuracy of MRD monitoring.24,31

This study describes a new ddPCR tool for MYD88L265P screening and MRD monitoring in WM.

Our results can be summarized as follows:

1.

ddPCR is a sensitive tool for MYD88L265P detection in WM and provides higher resolution compared to canonical ASqPCR;

2.

MYD88L265P ddPCR assay is particularly useful for reliable screening of low infiltrated WM specimens, such as unsorted BM or PB (routinely used in clinical practice);

3.

in treatment-naïve patients (TN) both BM and PB are informative when tested for MYD88L265P by ddPCR; however, in RE patients (mainly rituximab-exposed), PB has 1 log lower median MUT/WT ratio compared to BM, likely related to the efficacy of the anti-CD20 antibody in clearing the circulating tumor cells: therefore, for those patients, BM analysis should be preferred;

4.

MYD88L265P ddPCR assay can be reliably used for MRD purposes, being as informative as to the classical, standardized but less applicable, IGH-based MRD assay, not yet validated in WM;

(15)

13 5.

MYD88L265P ddPCR assay targeting ctDNA is very promising in identifying the mutation in less invasive tissues, like plasma or urine, and in samples from pre-treated patients.

The here described MYD88L265P ddPCR assay showed an overall mutation detection rate, on baseline unselected mononuclear cells samples, of 95.3% on BM and 71.2% on PB. These data might help to solve the still open debate on whether using sorted versus unsorted BM mononuclear cells to assess the MYD88L265P mutation36 and to extend the implementation of ddPCR mutation assay in routine diagnostic laboratories that do not routinely perform cells selection. Actually, only 40% of MYD88L265P cases can be detected by ASqPCR using unselected PB cells, with an overall sensitivity of 39.5% and specificity of 100%.37

In our study we observed that ddPCR assay, on unselected cells, greatly improves the rate of MYD88L265P detection compared to ASqPCR. Actually, the analysis on 74 paired BM/PB baseline samples showed an overall detection rate of 93% (69/74) on BM and 72% (53/74) on PB. Among 69 patients with detectable mutation on BM samples, the sensitivity for MYD88L265P mutation detection by ddPCR on paired PB samples was 77% (53/69). The highly sensitive results of MYD88L265P ddPCR assay on unsorted samples makes it ideal for diagnostic use in clinical routine, avoiding the costs and technical requirements of cell sorting. In addition, our data confirmed, as also reported by Xu L. et al37, that PB samples are suboptimal for the mutational screening in previously treated patients, evidence confirmed by the high rate of false negative results. Actually, the sensitivity of our assay in PB samples drops from 85% in TN to 47% in RE patients. Thus, to accurately identify the MYD88L265P WT status in pre-treated patients (strongly suggested for example before starting an expensive therapy with ibrutinib), a BM sample is essential.

(16)

14

This study shows that, besides its potential diagnostic role, MYD88L265P can effectively and easily be used for MRD monitoring in WM, achieving similar results to the less-applicable IGH-based MRD assay. Actually, allele burden levels monitoring can also provide insights on treatment effectiveness in a disease whose therapeutic scenario is rapidly changing, with many new and highly effective drugs.13 Of note, a report claimed the achievement of the first “molecular remission” in WM treated with carfilzomib.38 However, the deepness of molecular response does matter as well as the sensitivity of the assay used for MYD88L265P detection, being in that case quite modest (1.00E−03),6 compared to the newly developed flow cytometry assays39 and with the ddPCR tool described in this manuscript.

As we observed, the strength of agreement in the ddPCR vs ASqPCR comparison resulted lower in FU than in baseline samples. Further evaluations on large prospective patients series are needed to assess whether ddPCR could be useful for identification of false positive ASqPCR results, as recently demonstrated in a NGS/RQ-PCR comparison performed in acute lymphoblastic leukemia.40 Moreover, a methodological validation against IGH-based MRD detection and multiparamentric flow cytometry, as well as correlations with clinical impact, are eagerly awaited in this setting and are currently ongoing on external samples series.

The most innovative aspect of this study is the impact that ctDNA might have for MYD88L265P mutation detection and MRD monitoring. We showed that ctDNA mirrors the BM mutational burden much better than gDNA from PB at baseline. Moreover, our data suggest that ctDNA might be able to reflect the dynamic changes in tumor burden in response to treatment, with higher sensitivity than PB, particularly promising in pre-treated cases (Figures 3 and 4). Similar promising data have been obtained with ctDNA extracted from exosomes from PL and urine in a pivotal series of patients (unpublished), however further investigations, as well as technical optimization, are needed. Nevertheless, these

(17)

15

data foster the previously published experiences on the promising role of ctDNA analysis in lymphoproliferative disorders other than WM.41–50 Indeed, ctDNA represents an alternative, less invasive and “patient-friendly” tissue source for mutational analysis and eventually MRD, especially attractive for screening and monitoring of asymptomatic patients, such as MGUS cases (a MYD88L265P ddPCR screening on a large group of IgM-MGUS patients is currently ongoing). However, plasma collection needs particular care since blood collection tubes and drawing procedures, as well known, can bent the ctDNA stability.33-34 Pre-analytical standardized procedures are a prerequisite for clinical application and for consolidation of the promising potentials of ctDNA analysis.

Finally, we described the impact of different therapies on MRD clearance in WM (i.e. chlorambucil, rituximab mono, RCD (rituximab, cyclophosphamide, dexamethasone), FCR (fludarabine, cyclophosphamide, rituximab) and bendamustine-containing regimens: see online supplement). We acknowledge that among the limits of the present study there is the composite and retrospective nature of the patients series, moreover characterized by scanty outcome information. One of the main goal of this manuscript is the technical description of the MYD88L265P ddPCR assay relied on unsorted cells. The illustration of its wide potential applications, including those on liquid biopsy, it makes this approach potentially practical for implementation in routine diagnostic laboratories. We are aware that this assay represents an exploratory research, whose real advantages and predictive values need to be further validated in the context of prospective clinical trials. For this purpose, we are currently assessing MRD by ddPCR on gDNA and ctDNA in the context of a phase II prospective study for relapsed WM patients, sponsored by the Fondazione Italiana Linfomi, FIL (EudraCT Number: 2013-005129-22). Moreover, further efforts to

(18)

16

establish uniform guidelines regarding standardization procedures for samples collection and methodological validation are currently ongoing, both in national and European context.

In conclusion, our study highlights ddPCR as a feasible and highly sensitive assay for MYD88L265P mutational screening and MRD monitoring in WM, particularly in samples harboring low concentrations of circulating tumor cells. For this reason, plasmatic ctDNA represents a promising tissue source and might be an attractive, less invasive alternative to BM for MYD88L265P detection, also outside the WM scenario.

ACKNOWLEDGEMENTS

The authors would like to thank all the patients who participated in the study. We are grateful to Luca Arcaini, Alfredo Benso, Stefano Di Carlo, Angelo Fama, Paola Ghione, Idanna Innocenti, Luca Laurenti, Giacomo Loseto, Gianfranco Politano,Marzia Varettoni, Silvia Zibellini for their scientific advice and to Antonella Fiorillo and Sonia Perticone for administrative support.

(19)

17

REFERENCES

1. Treon SP, Xu L, Yang G, et al. MYD88 L265P somatic mutation in Waldenström’s macroglobulinemia. N Engl J Med. 2012;367(9):826–833.

2. Yang G, Zhou Y, Liu X, et al. A mutation in MYD88 (L265P) supports the survival of lymphoplasmacytic cells by activation of Bruton tyrosine kinase in Waldenström macroglobulinemia. Blood. 2013;122(7):1222–1232.

3. Campo E, Swerdlow SH, Harris NL, Pileri S, Stein H, Jaffe ES. The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications. Blood. 2011;117(19):5019–5032.

4. Jiménez C, Sebastián E, Chillón MC, et al. MYD88 L265P is a marker highly characteristic of, but not restricted to, Waldenström’s macroglobulinemia. Leukemia. 2013;27(8):1722–1728.

5. Poulain S, Roumier C, Decambron A, et al. MYD88 L265P mutation in Waldenstrom macroglobulinemia. Blood. 2013;121(22):4504–4511.

6. Xu L, Hunter ZR, Yang G, et al. MYD88 L265P in Waldenstrom macroglobulinemia, immunoglobulin M monoclonal gammopathy, and other B-cell lymphoproliferative disorders using conventional and quantitative allele-specific polymerase chain reaction. Blood. 2013;121(11):2051–2058.

7. Ngo VN, Young RM, Schmitz R, et al. Oncogenically active MYD88 mutations in human lymphoma. Nature. 2011;470(7332):115–119.

8. Varettoni M, Arcaini L, Zibellini S, et al. Prevalence and clinical significance of the MYD88 (L265P) somatic mutation in Waldenstrom’s macroglobulinemia and related

(20)

18

lymphoid neoplasms. Blood. 2013;121(13):2522–2528.

9. 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.

10. Treon SP, Cao Y, Xu L, Yang G, Liu X, Hunter ZR. Somatic mutations in MYD88 and CXCR4 are determinants of clinical presentation and overall survival in Waldenstrom macroglobulinemia. Blood. 2014;123(18):2791–2796.

11. Treon SP. How I treat Waldenström macroglobulinemia. Blood. 2009;114(12):2375– 2385.

12. Treon SP, Xu L, Hunter Z. MYD88 Mutations and Response to Ibrutinib in Waldenström’s Macroglobulinemia. N Engl J Med. 2015;373(6):584–586.

13. Leblond V, Kastritis E, Advani R, et al. Treatment recommendations from the Eighth International Workshop on Waldenström’s Macroglobulinemia. Blood. 2016;128(10):1321–1328.

14. Castillo JJ, Hunter ZR, Yang G, Argyropoulos K, Palomba ML, Treon SP. Future therapeutic options for patients with Waldenström macroglobulinemia. Best Pract Res Clin Haematol. 2016;29(2):206–215.

15. Abeykoon JP, Yanamandra U, Kapoor P. New developments in the management of Waldenström macroglobulinemia. Cancer Manag Res. 2017;973–983.

16. Hunter ZR, Yang G, Xu L, Liu X, Castillo JJ, Treon SP. Genomics, Signaling, and Treatment of Waldenström Macroglobulinemia. J Clin Oncol. 2017;35(9):994–1001.

17. Tedeschi A, Picardi P, Ferrero S, et al. Bendamustine and rituximab combination is safe and effective as salvage regimen in Waldenström macroglobulinemia. Leuk

(21)

19

Lymphoma. 2015;56(9):2637–2642.

18. Kyle RA. Long-term follow-up of IgM monoclonal gammopathy of undetermined significance. Blood. 2003;102(10):3759–3764.

19. Landgren O, Staudt L. MYD88 L265P somatic mutation in IgM MGUS. N Engl J Med. 2012;367(23):2255-2257.

20. Varettoni M, Zibellini S, Arcaini L, et al. MYD88 (L265P) mutation is an independent risk factor for progression in patients with IgM monoclonal gammopathy of undetermined significance. Blood. 2013;122(13):2284–2285.

21. Jiménez C, Chillón M del C, Balanzategui A, et al. Detection of MYD88 L265P Mutation by Real-Time Allele-Specific Oligonucleotide Polymerase Chain Reaction. Appl Immunohistochem Mol Morphol. 2014;22(10):768–773.

22. Gustine JN, Meid K, Hunter ZR, Xu L, Treon SP, Castillo JJ. MYD88 mutations can be used to identify malignant pleural effusions in Waldenström macroglobulinaemia. Br J Haematol. 2018;180(4):578-581.

23. Komatsubara KM, Sacher AG. Circulating Tumor DNA as a Liquid Biopsy: Current Clinical Applications and Future Directions. Oncology. 2017;31(8):618–627.

24. Hindson BJ, Ness KD, Masquelier DA, et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem. 2011;83(22):8604–8610.

25. Huggett JF, Whale A. Digital PCR as a novel technology and its potential implications for molecular diagnostics. Clin Chem. 2013;59(12):1691–1693.

(22)

20

immunoglobulin rearrangements for quantitative evaluation of minimal residual disease in multiple myeloma. Biol Blood Marrow Transplant. 2000;6(3):241–253.

27. van der Velden VHJ, Cazzaniga G, Schrauder A, et al. Analysis of minimal residual disease by Ig/TCR gene rearrangements: guidelines for interpretation of real-time quantitative PCR data. Leukemia. 2007;21(4):604–611.

28. Fleiss JL, Levin B, Paik MC. Statistical Methods for Rates and Proportions. Hoboken, NJ, USA. John Wiley & Sons, Inc. 2003.

29. Uchiyama Y, Nakashima M, Watanabe S, et al. Ultra–sensitive droplet digital PCR for detecting a low–prevalence somatic GNAQ mutation in Sturge–Weber syndrome. Sci Rep. 2016;6(1):22985.

30. Hunter ZR, Xu L, Yang G, et al. The genomic landscape of Waldenstrom macroglobulinemia is characterized by highly recurring MYD88 and WHIM-like CXCR4 mutations, and small somatic deletions associated with B-cell lymphomagenesis. Blood. 2014;123(11):1637–1646.

31. Drandi D, Kubiczkova-Besse L, Ferrero S, et al. Minimal Residual Disease Detection by Droplet Digital PCR in Multiple Myeloma, Mantle Cell Lymphoma, and Follicular Lymphoma. J Mol Diagnostics. 2015;17(6):652–660.

32. Petrikkos L, Kyrtsonis M-C, Roumelioti M, et al. Clonotypic analysis of immunoglobulin heavy chain sequences in patients with Waldenström’s macroglobulinemia: correlation with MYD88 L265P somatic mutation status, clinical features, and outcome. Biomed Res Int. 2014;2014:809103.

(23)

21

stability In K3EDTA, Streck, and CellSave blood collection tubes. Clin Biochem. 2016;49(18):1354–1360.

34. El Messaoudi S, Rolet F, Mouliere F, Thierry AR. Circulating cell free DNA: Preanalytical considerations. Clin Chim Acta. 2013;424:222–230.

35. Devonshire AS, Whale AS, Gutteridge A, et al. Towards standardisation of cell-free DNA measurement in plasma: controls for extraction efficiency, fragment size bias and quantification. Anal Bioanal Chem. 2014;406(26):6499–6512.

36. Gustine J, Meid K, Xu L, Hunter ZR, Castillo JJ, Treon SP. To select or not to select? The role of B-cell selection in determining the MYD88 mutation status in Waldenström Macroglobulinaemia. Br J Haematol. 2017;176(5):822–824.

37. Xu L, Hunter ZR, Yang G, et al. Detection of MYD88 L265P in peripheral blood of patients with Waldenström’s Macroglobulinemia and IgM monoclonal gammopathy of undetermined significance. Leukemia. 2014;28(8):1698–1704.

38. Treon SP, Tripsas CK, Meid K, et al. Carfilzomib, rituximab, and dexamethasone (CaRD) treatment offers a neuropathy-sparing approach for treating Waldenström's macroglobulinemia. Blood. 2014;124(4):503-510.

39. Paiva B, Montes MC, García-Sanz R, et al. Multiparameter flow cytometry for the identification of the Waldenström’s clone in IgM-MGUS and Waldenström’s Macroglobulinemia: new criteria for differential diagnosis and risk stratification. Leukemia. 2014;28(1):166–173.

40. Kotrova M, van der Velden VHJ, van Dongen JJM, et al. Next-generation sequencing indicates false-positive MRD results and better predicts prognosis after SCT in

(24)

22

patients with childhood ALL. Bone Marrow Transplant. 2017;52(7):962–968.

41. Kurtz DM, Green MR, Bratman S V, et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood. 2015;125(24):3679–3687.

42. 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.

43. Rossi D, Diop F, Spaccarotella E, et al. Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood. 2017;129(14):1947–1957.

44. Rustad EH, Coward E, Skytøen ER, et al. Monitoring multiple myeloma by quantification of recurrent mutations in serum. Haematologica. 2017;102(7):1266– 1272.

45. Sarkozy C, Huet S, Carlton VEH, et al. The prognostic value of clonal heterogeneity and quantitative assessment of plasma circulating clonal IG-VDJ sequences at diagnosis in patients with follicular lymphoma. Oncotarget. 2017;8(5):8765–8774.

46. Bruscaggin A, Spina V, Di Trani M, et al. Genotyping of Classical Hodgkin Lymphoma on the Liquid Biopsy. Hematol Oncol. 2017;3564–3565.

47. Camus V, Sarafan-Vasseur N, Bohers E, et al. Digital PCR for quantification of recurrent and potentially actionable somatic mutations in circulating free DNA from patients with diffuse large B-cell lymphoma. Leuk Lymphoma. 2016;57(9):2171–2179.

48. Alcaide M, Yu S, Bushell K, et al. Multiplex Droplet Digital PCR Quantification of Recurrent Somatic Mutations in Diffuse Large B-Cell and Follicular Lymphoma. Clin

(25)

23

Chem. 2016;62(9):1238–1247.

49. Hattori K, Sakata-Yanagimoto M, Suehara Y, et al. Clinical significance of disease-specific MYD88 mutations in circulating DNA in primary central nervous system lymphoma. Cancer Sci. 2018;109(1):225–230.

50. Hiemcke-Jiwa LS, Minnema MC, Radersma-van Loon JH, et al. The use of droplet digital PCR in liquid biopsies: A highly sensitive technique for MYD88 p.(L265P) detection in cerebrospinal fluid. Hematol Oncol. 2017 Dec 6. [Epub ahead of print].

(26)

24

Table 1: Patient’s clinical and biological features at baseline

Patients chacteristics Baseline samples (N= 148)

WM 133 (89,8%)

IgG-LPL 10 (6,7%)

IgM-MGUS 4 (2,8%)

IgM-LPL amyloid tumor 1 (0,7%)

Sex, F 51 (34%)

Median age (years, range) 67 (24-88)

Median Hemoglobin (g/dl, range) 11,5 (7,5-16,8) Median IgM (g/dl, range)

WM 2,397 (0,233-12,5)

MGUS 0,785 (0,492-2,252)

Median IgG (g/dl, range)

IgG-LPL 1,958 (0,916-3,447)

Median β2 microglobulin (mg/l, range) 2,62 (1-8,83) Median BM infiltration BM biopsy 40% (0-95%) BM flow cytometry 12% (0-87%) Organomegaly Splenomegaly 18 (12%) Adenopathies 29 (19,5%)

WM: Waldenström Macroglobulinemia; LPL: lymphoplasmacytic lymphoma; MGUS: monoclonal gammopathy of undetermined significance; F: female; BM: bone marrow.

(27)

25 Figure legend

Figure 1. MYD88L265P mutation at baseline is lower in PB compared to BM

A) WM patients show statistically significant differences in MUT/WT ratio between BM and PB samples (p<0.0001). The control group of healthy subjects and multiple myeloma (MM) patients show MUT/WT ratio below the LOD. Dashed red line shows the cut-off for mutational status. Symbols below the red line represent MYD88WT samples. B) MUT/WT ratio differences between relapsed (RE) and Naive to treatment (NT) patients, at baseline, is higher in PB compared to BM samples. C) MYD88L265P MUT/WT ratio at baseline in pared BM/PB samples from 55 NT and 19 RE patients, highlight the differences between biological specimens suggesting that for RE patients PB is less reliable than BM.

Figure 2. ddPCR sensitivity test and controls (MUT vs WT) plots examples

A) ddPCR assay measured on 10 fold dilution standard curve, shows a sensitivity of 0,0035% MUT (1 mutated copy in 30000 WT). B) Example of ddPCR result plot for the MYD88L265P control. C) Example of ddPCR result plot for the MYD88WT control.

Figure 3. MYD88L265P assay and IGH based approaches are superimposable

Comparison of MYD88 and IGH based MRD monitoring (express as copies of mutation in 1x105 cells) in 23 samples from 10 patients, show a good grade of correlation (r2=0,64).

Figure 4. cell tumor DNA from plasma mirrors the BM mutation level

MYD88L265P MUT/WT ratio in bone marrow (BM), peripheral blood (PB) and PLASMA (PL) paired samples from 32 patients show no statistically differences between BM and PL. Dashed red line shows the cut-off for mutational status. Symbols below the red line represent MYD88WT samples. (RE: relapsed; NT: Naive to treatment patients).

(28)

26 Figure 5. MYD88L265P can be used for disease monitoring

MYD88L265P monitored in 52 patients with at least 1 follow up (FU) sample.

(ctDNA: cell tumor DNA, RE: relapsed at baseline, NT: Naive to treatment at baseline, MRD : minimal residual disease, PL: plasma, BM: bone marrow, PB: peripheral blood).

(29)
(30)
(31)
(32)
(33)
(34)

SUPPLEMENTAL MATERIAL AND METHODS

Patients and samples collection

Three patient series were tested for MYD88L265P mutation by ddPCR. The local series of Torino (n=110) was used as discovery series, while the Salamanca and Pisa series (23 and 15 patients, respectively) were used for validation (Figure S1). Additionally, BM from 15 mantle cell lymphomas, 10 follicular lymphomas and PB from 10 chronic lymphocytic leukemia patients were tested. BM sample was filtered through a 1ml syringe, resuspend in erythrocytes lysis buffer (NH4Cl) (1:2 dilution) and leaved 15 minutes at room temperature (lying flat at dark), then centrifuged 10 minutes at 250xg at room temperature. The cell pellet was resuspended in 10-15 ml of NH4Cl and centrifuge again 10 minutes at 250xg at room temperature. The cell pellet was finally resuspended in PBS or 0.9% NaCl (q.s.), counted and dispensed as 5-10x106 cells, in each tube, centrifuged 1 minute at 13000xg and stored indefinitely, as dried pellets, at -80°C for further DNA extraction. Finally, 60 plasma and 32 urine samples were collected, as well. To ensure a good quality of cfDNA, plasma samples were collected in BCT tubes (Streck ) when coming from centers outside our hospital and in K3EDTA tubes, processed within 4 hours from draw, when coming from inside hospital (samples collection and storage are also listed in Table S1). Blood collected in Streck BCTs was shipped, stored and processed at room temperature. The first centrifugation was conducted at 1300×g for 13 min at 4 °C for K3EDTA tubes (while BCTs at room temperature), after which the plasma was carefully removed and transferred to a new tube. The second centrifugation was performed at 1800×g for 10 min. The resultant plasma was collected and stored at −80 °C until analysis. Plasma samples were thawed and centrifuged at 13000 ×g for 3 min before cfDNA extraction.

(35)

Figure S1: Overview of patients and samples characteristics, collected for MYD88L265P mutation detection by ddPCR. BM:bone marrow, PB: peripheral blood, NT: naïve to treatment at baseline, RE: relapsed at baseline.

Nucleic acid extraction

Genomic (gDNA) from 5x106 cells and cell-free DNA (cfDNA) from 1 ml of plasma, was extracted by Maxwell automatic system (MaxWell RSC, Promega), following the manufacturer recommendations. To ensure extraction quality and no contamination from gDNA, selected cfDNA samples, with different amount of WT copies, were tested with the Specimen control size ladder kit (InVivoScribe Technologies). Moreover, the amount of cfDNA was also evaluated by RnaseP gene.

(36)

ddPCR assays for MYD88L265P mutation detection

Mutation detection assay was designed as reported in Figure S2A. A single set of primers

(Forward FP 5'-CCTTGGCTTGCAGGT-3' and Reverse RP

5'-TCTTTCTTCATTGCCTTGT-3') was combined with two competitive probes in two assays (CSTM DDPCR HEX/FAM ASSAY BIO-RAD), one for MYD88L265P mutation (MUT 5'-TGGGGATCGGTCGC-3') labeled with FAM and one for MYD88L265P wild type (WT 5'-TGGGGATCAGTCGCTT-3') labeled with HEX. At first, the ddPCR assay was optimized for DNA amount and PCR conditions using the QX100 Droplet Digital PCR system (Bio-Rad Laboratories, Hercules, CA, USA). Briefly, for each replicate, 11 l of 2X ddPCR Supermix for Probes with no dUTP (Bio-Rad Laboratories), 1.1 l of each 20X mutation detection assay and 5.5 l of gDNA (20 ng/l) or cfDNA were mixed in a total 22 l reaction volume. Droplets were generated, by a QX100 droplet generator device, from 20 l of the reaction mix, and end-point PCR was performed on a T100 Thermal Cycler (Bio-Rad Laboratories) at following conditions: 95°C for 10 minutes, 40 cycles of 94°C for 30 seconds, 55°C for 1 minute followed by 98°C for 10 minutes. Ramp rate was set at 2.5°C/second. PCR products were loaded into the QX100 droplet reader and analyzed by QuantaSoft v1.6.6.0320 (Bio-Rad Laboratories). Samples were tested in triplicate and results expressed as merge of wells. ddPCR gate setting was performed based on the positive control sample. The cut-off for mutation was set based on the highest MYD88L265P level detected within the control group of healthy samples and is indicated in figures as a dashed line. dMIQE guidelines (Minimum Information for Publication of Quantitative Digital PCR Experiments) for ddPCR experiments are listed in Table S1.1

ASOqPCR assay for tumor-specific IGH-VDJ rearrangement

Patient-specific IGH-VDJ rearrangements were amplified and directly sequenced from diagnostic gDNA. Sequences were analyzed using the IMGT/V-QUEST tool [http://imgt.org].2,3 and patient-specific ASO primers and consensus probes were designed, as previously described (Figure S2B) 4. IGH-based MRD analysis was performed according to the Euro-MRD guidelines5.

(37)

Figure S2: PCR strategies for A) MYD88L265P mutation detection by ddPCR and ASqPCR and B) IGH rearrangement quantification by ddPCR. ASO: allele specific oligonucleotide (clone specific primers), MUT: mutated, WT: wildtype, SbrG: Sybergreen dye.

(38)

ITEM IMPORTANCE INCLUDED COMMENTS

Experimental Design

Definition of experimental

and control groups E Y

Number within each group E Y

Assay carried out by core

lab or investigator's lab? D Investigator's lab

Power analysis D Y

Sample

Description E Y

Volume or mass of

sample processed E 10 ml of peripheral blood (PB) and 5ml of bone marrow (BM)

Microdissection or

macrodissection E n n/a

Processing procedure E

PB were centrifuged 13 minutes at 1300Xg at +4°C, plasma was centrifuged 10 minutes at 1800Xg at room temperature. Urine sample were

centrifugated 10 minutes at 200Xg at room temperature. If frozen - how and how

quickly? E

Plasma samples were collected on BTC tubes (StrecK) when came from outside hospital and within 4 hours from draw when came from inside

hospital If fixed - with what, how

quickly? E n n/a

Sample storage conditions and duration (especially for FFPE samples)

E plasma and urine were stored in 1 ml aliquotes, PB and BM cells in dryed

pellets both at -80°C, Nucleic Acid Extraction Quantification-instrument/method E Nanodrop 2000 Storage conditions: temperature, concentration, duration, buffer

E gDNA and cfDNA were stored at -20°C

DNA or RNA quantification E after extraction and dilution

Quality/integrity,

instrument/method E Y

Template structural

information E not performed

Template modificcation (digestion, sonication, preamplification, etc.)

E not performed

Template treatment (initial heating or chemical debnaturation)

E not performed

Inibhition dilution or spike E n n/a

DNA contamination

assessment of RNA sample E n n/a

Details od Dnase treatment

where performed E n n/a

Manufacturer of reagents

used and catalogue number D n n/a

Storage of nucleic acid: temperature, concentration, duration, buffer

E cfDNA from plasma in 60 uL, urine in 40 ul, gDNA in 100 ul of elution

buffers ddPCR Target Information Sequence accession number E n n/a

Amplicon location D n see RTPrimerDB (ID:1529)

Amplicon length E 65-mer

In silico specificity

screen (BLAST, etc) E n n/a

Pseudogenes, retropseudogenes or other homologs?

D n n/a

Sequence alignment D n n/a

Secondary structure analysis of amplicon and GC content

D n n/a

Location of each primer by

exon or intron (if applicable) E n n/a

Which splice variants

are targeted? E n n/a

(39)

ddPCR Oligo nucleotides

Primer sequences and/or

amplicon context sequence E Y

RTPrimerDB Identification

Number D n n/a

Probe sequences D Y

Location and identity of any

modifications E Y

Manufacturer of

oligonucleotides D Integrated DNA Technologies (IDT), Coralville, IA, USA

Purification method D Desalting

ddPCR Protocol

Complete reaction

conditions E Y

Reaction volume and

amount of cDNA/DNA E Y

Primer, (probe), Mg2+

and dNTP concentrations E Y (ONLY FOR PRIMERS AND PROBE)

Polymerase identity and

concentration E n n/a

Buffer/kit catalougue no.

and manufacturer E

2X ddPCR Supermix for Probes (No dUTP) (cat. 186-3024), ddPCR droplet generation oil for probes (cat. 186-3005), ddPCR droplet reader oil

(cat. 186-3004 ) (Bio-Rad Laboratories, Hercules, CA, USA) Exact chemical

constitution of the buffer D n n/a

Additives (SYBR Green

I, DMSO, etc.) E n n/a

Plates/tubes catalog no and

Manufacturer D

DG8 cartridges (cat. 186-4008), DG8 gaskets (cat. 186-3009), pierceable foil heat seals (cat. 181-4040),

Complete thermocycling

parameters E Y

Reaction setup D Y

Gravimentric or volumetric

dilutions D Y

total PCR reaction volume

prepared D Y

Partition number E > 9000 UP TO 22000

Individual partition volume E Y

Total volume of partitions measured (effective reaction size)

E 40L

Partition volume

varianca/SD D n

Comprehensive details and

appropriate use of controls E Y

Manufacturer of dPCR

instrument E Y

ddPCR validation

Optimization data for the

assay E Y

Specificity E Y

Limit of detection of

calibration control E Y

if multiplexing, comparison

(40)

ITEM IMPORTANCE INCLUDED COMMENTS

Data Analysis

Mean copies per partition

(Delta or equivalent) E n n/a

dPCR analysis program

(source, version) E Y

Outlier identification and

disposition E n n/a

Results of NTCs E All reactions shown negative NTC

Examples of positive and negative experimental results as supplemental data

E Y

Justification of number and

choice of reference genes E n n/a

Description of normalization

method E n n/a

Number and concordance

of biological replicates D n n/a

Number and stage of

technical replicates E Y Repeatability (intra-assay variation) E Y Reproducibility (inter-assay variation) D Y Experimental variance or CI E Y

Statistical methods used for

analysis E Y

Data submission using

RDML D n n/a

Table S1. Checklist for (E) essential and (D) desirable dMIQE information useful for ddPCR experiments.

(41)

Supplemental results

ddPCR limit of detection

LOD (0.035%) was defined using a statistical method based on binomial distribution, as previously reported6. However, we observed consistently positive replicates at one log lower dilution point of the standard curve 0,0035% (1 copy of MUT out of 30000 WT) (Table S2). In order to evaluate the reliability of the calculated LOD, based on the concept that the detection limit is a function of both signal strength and stability, we tested the reliability of 0.035% dilution point performing additional experiments. Ninety replicates were tested, to verify the reproducibility, for the 0,0035% and for the half log higher 0,018% dilution point (5 MUT copies in 30000 WT). While for 0.035% al replicates were positive, the level of reproducibility was 54/90 and 71/90 positive wells, respectively for 0,0035% and 0.018% dilution point (Table S2) showing inconsistently positive or negative replicates and confirming that the above calculated LOD and the experimentally detected cut-off, arising from the control group, are both reliable.

(42)

Table S2. MUT/WT ratio detected by ddPCR on 10 fold dilution standard curve. From

35% to 0.0035 % corresponding to 10500, 1050, 105, 10.5 and 1 mutated copy numbers. Each dilution point was evaluated in triplicate. Reproducibility for 1 copy (0,0035%) and 4

copies (0,018%) on 90 replicated is also reported.

Comparison of MYD88L265P ddPCR versus ASqPCR assays

(43)

Figure S4: ddPCR versus AsqPCR. Plots, for both methods, reported from two patients

(44)

MYD88L265P ddPCR on ctDNA

Figure S5: MYD88L265P MUT/WT ratio in PB/ctDNA from PLASMA (PL) paired samples

from 60 patients. Dashed line shows the negativity cut-off depicted based on MUT/WT ratio within the control samples group.

(45)

Figure S6: MYD88L265P MUT/WT ratio in BM-PLASMA-PB paired samples from 32 patients. Dashed line shows the negativity cut-off depicted based on MUT/WT ratio within the control samples group.

(46)

MRD monitoring in WM by MYD88L265P ddPCR

WM 45 (86,6%)

IgG LPL 6 (11,5%)

LPL amyloid tumor 1 (1,9%)

Sex, F 28 (53,8%)

Median age (years, range) 67 (24-81)

Median β2 microglobulin (mg/l, range) 2,61 (1-6)

BM biopsy 40% (0-95%) BM flow cytometry 10% (0-44%) Splenomegaly 7 (13,4%) Adenopathies 13 (25%) W&W 11 (21,2%) RTX single agent 2 (3,8%) Chlorambucil 6 (11,5%) RCD-like 13 (25%) FCR-like 3 (5,8%) RBendamustine-based 6 (11,5%) Bortezomib-based 8 (15,4%) Others 3 (5,8%)

(N= 52) Selected patients chacteristics

Median BM infiltration

Organomegaly

First line treatment

Table S4. Abbreviations: WM, Waldenström Macroglobulinemia; LPL, lymphoplasmacytic

lymphoma; F, female; BM, bone marrow; W&W, watch & wait; RTX, rituximab; RCD, rituximab-cyclophosfamide-dexamethasone;

(47)

Figure S7A. MYD88L265P mutation monitoring in NT patients, after Chemo-therapy (chlorambucil or rituximab alone as first therapy).

(48)

Figure S7B. MYD88L265P mutation monitoring in NT patients after standard R-Chemo-therapy. RCD: Rituximab, Cyclophosphamide, Dexamethasone; FCR: Fludarabine, Cyclophosphamide, Rituximab.

(49)

Figure S7C. MYD88L265P mutation monitoring after Rituximab-Bendamustine based therapy.

(50)

References

1. Huggett JF, Foy CA, Benes V, Emslie K, Garson JA, Haynes R, et al. The digital MIQE guidelines: Minimum Information for Publication of Quantitative Digital PCR Experiments. Clin Chem 2013; 59(6): 892-902.

2. Giudicelli V, Brochet X, Lefranc M-P. IMGT/V-QUEST: IMGT standardized analysis of the immunoglobulin (IG) and T cell receptor (TR) nucleotide sequences. Cold Spring Harb Protoc. 2011;2011:695–715.

3. Brochet X, Lefranc M-P, Giudicelli V. IMGT/V-QUEST: the highly customized and integrated system for IG and TR standardized V-J and V-D-J sequence analysis. Nucleic Acids Res. 2008;36: 503-508.

4. Ladetto M, Donovan JW, Harig S, et al. Real-Time polymerase chain reaction of immunoglobulin rearrangements for quantitative evaluation of minimal residual disease in multiple myeloma. Biol Blood Marrow Transplant 2000;6(3):241–253.

5. van der Velden VHJ, Cazzaniga G, Schrauder A, et al. Analysis of minimal residual disease by Ig/TCR gene rearrangements: guidelines for interpretation of real-time

quantitative PCR data. Leukemia 2007;21(4):604–611.

6. Uchiyama Y, Nakashima M, Watanabe S, et al. Ultra–sensitive droplet digital PCR for detecting a low–prevalence somatic GNAQ mutation in Sturge–Weber syndrome. Sci Rep 2016;6(1):22985.

Riferimenti

Documenti correlati

Distribution of the marine and transitional terraces (Holocene) and geomorphological map of the coastal area between Punta Paulo and Porvenir – Tierra del Fuego, Straits of Magellan

Use of all other works requires consent of the right holder (author or publisher) if not exempted from copyright protection by the applicable law.. Taylor M.E.,

underlined that the most effective way to correct vitamin D deficiency in elderly subjects is to administer a personalized loading dose based on body weight and baseline 25OHD

I parametri relativi alla motilità, alla velocità, alla vi- talità ed alla morfologia sono risultati essere in- fluenzati, nel nostro caso, non tanto dalla razza quanto invece

Following reports of human cases of Lyme borreliosis from the Ossola Valley, a mountainous area of Piemonte, north-western Italy, the abundance and altitudinal

We prove the existence of orbitally stable ground states to NLS with a partial confinement together with qualitative and symmetry properties1. This result is obtained for

Measured performance is evaluated with respect to several system parameters: tag number and typology, tag reciprocal distance and orientation, trajectory and speed of the

Si faccia quindi attenzione: nel punto dello svolgimento delle Anmerkungen in cui ci troviamo, alla fine, cioè, del secondo paragrafo, Strauss sostiene che il «ritorno» a Hobbes,