kazuko ino · web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 conditioning regimen...

50
Title page Clinical utility of WT1 monitoring in patients with myeloid malignancy and prior allogenic hematopoietic stem cell transplantation Kazuko Ino 1,2) , Shigeo Fuji 1) , Kinuko Tajima 1) , Takashi Tanaka 1) , Keiji Okinaka 1) , Yoshihiro Inamoto 1) , Saiko Kurosawa 1) , Sung-Won Kim 1) , Naoyuki Katayama 2) , Takahiro Fukuda 1) 1) Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan. 2) Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan. Corresponding author: Shigeo Fuji, M.D. Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan 5-1-1, Tsukiji, Chuo-Ku, Tokyo 104-0045, Japan. TEL: +81-3-3542-2511 1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20

Upload: others

Post on 02-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Title page

Clinical utility of WT1 monitoring in patients with myeloid malignancy and prior allogenic

hematopoietic stem cell transplantation

Kazuko Ino1,2), Shigeo Fuji1), Kinuko Tajima1), Takashi Tanaka1), Keiji Okinaka1), Yoshihiro

Inamoto1), Saiko Kurosawa1), Sung-Won Kim1), Naoyuki Katayama2), Takahiro Fukuda1)

1) Department of Hematopoietic Stem Cell Transplantation, National Cancer Center

Hospital, Tokyo, Japan.

2) Department of Hematology and Oncology, Mie University Graduate School of Medicine,

Tsu, Japan.

Corresponding author:

Shigeo Fuji, M.D.

Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital,

Tokyo, Japan

5-1-1, Tsukiji, Chuo-Ku, Tokyo 104-0045, Japan.

TEL: +81-3-3542-2511

FAX: +81-3-3542-3815

E-mail: [email protected]

1

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 2: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Running title: WT1 as MRD in allo-HSCT

Key words: allogeneic hematopoietic stem cell transplantation, myeloid malignancy,

minimal residual disease.

Word count: abstract 197; main body 24593120

Abstract

Although allogeneic hematopoietic stem cell transplantation (allo-HSCT) is one of the

standard treatments for myeloid malignancy, relapse remains a major obstacle to cure.

Early detection of relapse by monitoring of minimal residual disease (MRD) may enable us

to intervene preemptively and potentially prevent overt relapse.

WT1 is well known as a panleukemic marker. We retrospectively examined serially

monitored WT1 levels of peripheral blood in 98 patients (84 with acute myeloid leukemia

and 14 with myelodysplastic syndrome). At the time of allo-HSCT, 49 patients (50%) were

in CR. Patients were divided into three groups according to WT1 levels (<50 copy/gRNA,

50-500 copy/gRNA and >500 copy/gRNA). The cumulative incidence of relapse (CIR)

and overall survival (OS) differed statistically according to the WT1 levels before allo-HSCT

and at days 30 and 60 after allo-HSCT. In multivariate analysis, WT1 >500 copy/gRNA

2

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 3: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

before and at day 60 after allo-HSCT, and WT1 >50 copy/gRNA at day 30 were correlated

with CIR. Moreover, WT1 >500 copy/gRNA at day 60 after allo-HSCT was only correlated

with worse OS. Our data suggest that serial monitoring of WT1 levels in peripheral blood

may be useful for MRD monitoring and as a predictor of hematological relapse in allo-

HSCT.

Introduction

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has recently become a

standard treatment for patients with high-risk myeloid malignancies. A number of patients

are cured after allo-HSCT. For other patients and their transplant teams, however, relapse

after allo-HSCT remains a major obstacle to treatment success. Preemptive intervention in

patients with an impending relapse is an attractive option, and identification of such patients

requires monitoring techniques to detect minimal residual disease (MRD).

Some reports have indicated that in patients with acute myeloid leukemia (AML),

detection of MRD after induction chemotherapy and before allo-HSCT correlates with the

risk of relapse after allo-HSCT (ref. 1-9).

In patients with myeloid malignancies characterized by expression of a chimeric fusion

gene such as PML/RAR, AML1/MTG8, or CBF/MYH11, this expression can be used as

the MRD parameter. On the other hand, a significant proportion of patients with myeloid

malignancies do not have such markers, which makes the monitoring of MRD difficult.

3

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 4: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Wilms’ tumor 1 (WT1) was originally identified as a tumor suppressor gene in Wilms’ tumor,

a pediatric renal cancer (ref. 10, 11). WT1 is also overexpressed in many myeloid

malignancies and can be used for monitoring MRD in patients’ peripheral blood (PB) (ref. 2,

4, 9, 12-21). Although the superiority of WT1 level in PB in comparison to that in bone

marrow has not yet been established, the sampling of PB is obviously preferable in clinical

practice. Thus, we assessed the importance of WT1 level in PB only in this study. At our

center, WT1 levels of PB was measured as MRD monitoring routinely. However, clinical

decisions based on the results of WT1 level relies mostly on the decisions of physicians as

the threshold has not been well established. At our institute, patients who underwent allo-

HSCT were followed monthly to monitor WT1 levels in PB. In this report, we retrospectively

assessed the utility of WT1 monitoring and the correlation between WT1 levels and clinical

outcomes.

Patients and methods

Study population and WT1 analysis

Our study included 149 adult patients who had myeloid malignancies and received their

first allo-HSCT at the National Cancer Center Hospital, Tokyo, Japan, from April 2010 to

August 2014. We excluded 47 patients without data of WT1 levels, 3 patients with isolated

myeloid sarcoma, and 1 patient with myeloid NK precursor acute leukemia. We analyzed

the detailed clinical features of the remaining 98 patients. WT1 levels in PB were

4

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 5: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

determined before and 30 and 60 days after allo-HSCT, using the WT1 mRNA Assay Kit II

“new kit” (Otsuka Pharmaceutical Co., Ltd., Tokyo, Japan) as previously reported (ref. 22).

The WT1 mRNA expression levels were calculated by multiplying the value obtained by

dividing the measured WT1 mRNA by the measured value of GAPDH mRNA (number of

WT1 mRNA copies per copy of GAPDH mRNA) with the mean GAPDH mRNA

measurement value per 1 g of RNA (2.7 x 107 copies/g RNA) based on independent

tests in healthy adults. The method to calculate WT1 mRNA expression is shown below. A

unit of WT1 mRNA expression was prescribed as copies/g RNA.

WT1 mRNA expression (copies/g RNA) = [Measured value of WT1 mRNA (copies/mL)

/Measured value of GAPDH mRNA (copies/mL)] x 2.7 x 107 (copies/g RNA)*

*2.7 x 107 (copies/gRNA): mean GAPDH mRNA measurement value per 1 g of RNA in

PB of healthy adult.

The IRB of National Cancer Center approved this study. In this study, complete remission

(CR) and relapse were defined as hematological. These data were analyzed as of March

2015. The median follow-up time of survivors, defined as the time from allo-HSCT to last

observation, was 775 days.

MRD detection

WT1 levels in PB were serially monitored. Patients were divided into three groups

according to WT1 levels, as follows: group 1, <50 copy/gRNA; group 2, 50-500

5

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 6: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

copy/gRNA; and group 3, >500 copy/gRNA. The receiver operating characteristic (ROC)

curve for the prediction of hematological relapse indicated that the best cutoff values at

each time point were as follows: WT1 600 copy/gRNA before allo-HSCT, 87 copy/gRNA

at day 30 after allo-HSCT, and 120 copy/gRNA at day 60 after allo-HSCT. We further

analyzed the cutoff value of WT1 500 copy/gRNA. WT1 50 copy/gRNA was the detection

limit in this analysis. The MRD data was obtained before and at approximately days 30 and

60 after allo-HSCT. Patients were classified according to their MRD values.

Statistical methods

To estimate the probabilities of cumulative incidence of relapse (CIR) and overall survival

(OS), the observation time was calculated from the MRD examination to the event date of

the last follow-up. Evaluation of CIR was performed with the Fine and Gray model and

Gray’s test, and non-relapse mortality (NRM) was considered a competing risk for relapse.

NRM was defined as death without prior relapse. The probabilities of OS were estimated

with Kaplan-Meier (KM) statistics. The log-rank test was used for comparisons. Cox

proportional hazards regression model analysis was conducted to identify factors affecting

an adverse subsequent event. Covariates were further investigated in a multivariate Cox

proportional hazard model based on stepwise selection strategy, and the main effect

variable of MRD was held in all steps of model building. The corresponding hazard ratios

(HRs) and their 95% confidence intervals (95%CIs) were calculated.

6

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 7: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

All statistical analyses were performed with EZR software (ref. 23), which is a graphical

user interface for R. More precisely, it is a modified version of R commander designed to

add statistical functions frequently used in biostatistics.

Results

Patient characteristics

A total of 98 patients were included in this retrospective study; their detailed characteristics

are shown in Table 1. The median age was 46.5 years (range, 18-68 years). Eighty-four

patients (85.7%) had AML and 14 (14.3%) had myelodysplastic syndrome (MDS). At the

time of allo-HSCT, 49 patients (50%) achieved CR. The median follow-up period among

survivors was 775 days (range, 61-1739 days) after allo-HSCT. Cytogenetic risk was good

in 47 patients (47.9%), intermediate in 27 patients (27.6%), and poor in 20 patients

(20.4%). Of the 47 patients with good cytogenetic risk, 4 were AML1/MTG8 positive, 1 was

CBF/MYH11 positive, and 1 was PML/RAR positive. All patients with good cytogenetic

risk suffered from relapsed disease after first complete remission or primary induction

failure. Thus, it would be reasonable to consider those patients as a candidate for all-HSCT.

This karyotype analysis was based on the International Prognostic Scoring System (ref.

24). Twenty-seven patients (27.5%) received allo-HSCT from related donors and the others

from unrelated donors. In terms of HLA disparity, 23 patients (23.5%) had 1 locus mismatch

and 30 patients (30.6%) had >2-loci mismatches. The conditioning regimens were as

7

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 8: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

follows: 28 patients (28.6%) received total body irradiation / cyclophosphamide (CY), 32

patients (32.7%) received busulfan (Bu) / CY, 12 patients (12.2%) received fludarabine

(Flu) / Bu 3.2 mg/kg/day for 4 days as a myeloablative conditioning regimen, and 26

patients (26.5%) received Flu / Bu 3.2 mg/kg/day for 2 days as a reduced-intensity

conditioning regimen. Myeloablative conditioning regimen was used in 72 patients (73.5%).

Monitoring of WT1 levels and the relationship to clinical outcomes

In this study, we retrospectively analyzed the data of WT1 in PB. These results are shown

in Figure 1. Most patients with WT1 levels <500 copy/gRNA before allo-HSCT maintained

WT1 levels <50 copy/gRNA after allo-HSCT. In contrast, of the patients with WT1 levels

>500 copy/gRNA, about one-third demonstrated WT1 levels above 50 copy/gRNA after

allo-HSCT.

CIR and OS differed statistically (P<0.01) according to the WT1 levels before allo-HSCT

(Figure 2a and b). The 2-year relapse rates were 21% (95%CI 9-36%) in the WT1 <50

copy/gRNA group, 23% (95%CI 5-48%) in the WT1 50-500 copy/gRNA group, and 49%

(95%CI 33-64%) in the WT1 >500 copy/gRNA group (Figure 2a). The 2-year OS rates

were 90% (95%CI: 73-96%) in the WT1 <50 copy/gRNA group, 79% (95%CI 36-94%) in

the WT1 50-500 copy/gRNA group, and 52% (95%CI 34-67%) in the WT1 >500

copy/gRNA group (Figure 2b).

CIR and OS differed statistically (P<0.01) according to the WT1 levels at days 30 and

8

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 9: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

60 after allo-HSCT (Figure 2c-f). Grouped according to the WT1 level at day 30 after allo-

HSCT, the 2-year relapse rates were 27% (95%CI 18-38%) in the WT1 <50 copy/gRNA

group, 38% (95%CI 13-63%) in the WT1 50-500 copy/gRNA group, and 83% (95%CI 8-

98%) in the WT1 >500 copy/gRNA group (Figure 2c). Grouped according to the WT1

level at day 30 after allo-HSCT, the 2-year OS rates were 76% (95%CI 64-84%) in the WT1

<50 copy/gRNA group, 82% (95%CI 46-95%) in the WT1 50-500 copy/gRNA group, and

42% (95%CI 9-73%) in the WT1 >500 copy/gRNA group (Figure 2d). Grouped according

to the WT1 level at day 60 after allo-HSCT, the 2-year relapse rates were 21% (95%CI 13-

31%) in the WT1 <50 copy/gRNA group, 50% (95%CI 12-79%) in the WT1 50-500

copy/gRNA group, and not available in the WT1 >500 copy/gRNA group (Figure 2e).

Grouped according to the WT1 level at day 60 after allo-HSCT, the 2-year OS rates were

85% (95%CI 74-91%) in the WT1 <50 copy/gRNA group, 55% (95%CI 20-80%) in the

WT1 50-500 copy/gRNA group, and not available in the WT1 >500 copy/gRNA group

(Figure 2f).

Univariate and multivariate analysis for relapse

In univariate analysis, sex (vs. male; female, HR 0.40, 95%CI 0.21-0.78, P<0.01), status of

disease at the time of allo-HSCT (vs. CR; non-CR, HR 5.16, 95%CI 2.46-10.82, P<0.01),

and karyotype (vs. good; poor, HR 2.89, 95%CI 1.53-5.49, P<0.01) were associated with

an increased risk of CIR. In terms of MRD before allo-HSCT, WT1 >500 copy/gRNA was

9

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 10: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

associated with an increased risk of CIR (vs. WT1 <50 copy/gRNA; WT1 >500

copy/gRNA, HR 4.22, 95%CI 2.04-8.76, P<0.01). In terms of MRD at day 30, WT1 >500

copy/gRNA was associated with an increased risk of CIR (vs. WT1 <50 copy/gRNA at

day 30; WT1 >500 copy/gRNA at day 30; HR 7.43, 95%CI 2.17-25.43, P<0.01). In terms

of MRD at day 60, WT1 >500 copy/gRNA was associated with an increased risk of CIR

(vs. WT1 <50 copy/gRNA at day 60; WT1 >500 copy/gRNA at day 60; HR 24.73, 95%CI

8.32-73.51, P<0.01, Table 2).

In multivariate analysis, WT1 >500 copy/gRNA before allo-HSCT was associated with

an increased risk of CIR (vs. WT1 <50 copy/gRNA; WT1 >500 copy/gRNA, HR 3.00,

95%CI 1.20-7.49, P=0.02) (Table 3). In terms of MRD at day 30, WT1 50-500 copy/gRNA

and >500 copy/gRNA were associated with an increased risk of CIR (vs. WT1 <50

copy/gRNA at day 30; WT1 50-500 copy/gRNA at day 30, HR 2.98, 95%CI 1.19-7.47,

P=0.02; WT1 >500 copy/gRNA at day 30, HR 7.86, 95%CI 2.28-27.12, P<0.01) (Table 3).

In terms of MRD at day 60, WT1 >500 copy/gRNA was associated with an increased risk

of CIR (vs. WT1 <50 copy/gRNA at day 60; WT1 >500 copy/gRNA at day 60, HR 15.90,

95%CI 4.22-59.92, P<0.01) (Table 3).

Univariate and multivariate analysis of OS

In univariate analysis, the disease status at the time of allo-HSCT (vs. CR; non-CR, HR

6.08, 95%CI 2.28-16.21, P<0.01), and karyotype (vs. good; poor, HR 3.08, 95%CI 1.35-

10

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 11: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

7.02, P<0.01) were adverse prognostic factors for OS. In terms of MRD before allo-HSCT,

WT1 >500 copy/gRNA was an adverse prognostic factor for OS (vs. WT1 <50

copy/gRNA; WT1 >500 copy/gRNA, HR 5.41, 95%CI 2.13-13.76, P<0.01). In terms of

MRD at day 30, WT1 >500 copy/gRNA was an adverse prognostic factor for OS (vs. WT1

<50 copy/gRNA at day 30; WT1 >500 copy/gRNA at day 30, HR 5.11, 95%CI 1.70-15.34,

P<0.01). In terms of MRD at day 60, WT1 50-500 copy/gRNA and >500 copy/gRNA were

adverse prognostic factors for OS (vs. WT1 <50 copy/gRNA at day 60; WT1 50-500

copy/gRNA at day 60, HR 3.20, 95%CI 1.04-9.87, P=0.04; WT-1 >500 copy/gRNA at day

60, HR 25.78, 95%CI 6.62-100.40, P<0.01) (Table 4).

In multivariate analysis, WT1 >500 copy/gRNA at day 60 of allo-HSCT was the only

adverse prognostic factor for OS (vs. WT1 <50 copy/gRNA at day 60; HR 13.94, 95%CI

3.59-54.11, P<0.01) (Table 5). In contrast, WT1 levels before and at day 30 after allo-HSCT

were not significant risk factors for OS (Table 5).

Impact of conditioning intensity on the kinetics of WT1 level after allo-HSCT

With regard to the conditioning intensity, a univariate analysis showed a trend toward

higher rate of MRD negativity in patients who received a myeloablative conditioning

regimen (80.5% at day 30, 84.3% at day 60) in comparison to those who received a non-

myeloablative conditioning regimen (76.9% at day 30, 75.0% at day 60) (Supplemental

Table 1). However, conditioning intensity was not a significant variable in multivariate

11

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 12: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

analysis.

Subgroup analysis stratified based on the pretransplant disease status

We added the subgroup analysis stratified based on the pretransplant disease status

(Supplemental Figure 1, 2). In terms of patients in hematological CR with WT1 level >500

copy/gRNA, the number of patients was limited (n=6), although the cumulative incidence

of relapse seems to be high. The study population was small, further studies were needed.

Discussion

In this study, we showed that serial monitoring of WT1 in PB before and after allo-HSCT

was a useful method to estimate the risk of hematological relapse in patients with myeloid

malignancies, as elevation of WT1 level was significantly associated with an increased risk

of subsequent hematological relapse.

In multivariate analysis, WT1 >500 copy/gRNA before and at day 60 after allo-HSCT,

and WT1 >50 copy/gRNA at day 30 after allo-HSCT were significantly associated with an

increased risk of CIR. Moreover, WT1 >500 copy/gRNA at day 60 after allo-HSCT was the

only significant prognostic risk factor for OS. The assessment and monitoring of MRD after

allo-HSCT are of crucial importance in patients with myeloid malignancies, as they can

enable us to determine transplantation efficacy and to achieve early diagnosis of relapse.

We had data of WT1 levels at day 90 and day 180 after HSCT (Supplemental table 2).

12

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 13: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

At day 90, 74 patients were <50 copy/gRNA, 7 patients were 50-500 copy/gRNA and 2

patients were >500 copy/gRNA. In these 9 patients of MRD positivity, 6 patients (66.7%)

had subsequent hematological relapse. Meanwhile, at day 180, 69 patients were <50

copy/gRNA, 2 patients were 50-500 copy/gRNA and 3 patients were >500 copy/gRNA.

In these 5 patients of MRD positivity, all patients (100%) had subsequent hematological

relapse. Although the number of evaluable patients decreased at later time point, long-term

monitoring of WT1 level in PB might be useful to assess the risk of subsequent

hematological relapse. On the contrary, MRD detection of day 30 may be uninformative

because of recovering hematopoiesis after allo-HSCT. In this study, the possibility of MRD

value of WT1 at day 30 was suggested. Further studies will be needed to detail analysis at

this time point.

WT1 elevation was eventually detected in 40 patients (Figure 3), of whom 27 had

subsequent hematological relapse. Four of these 27 patients were alive after second allo-

HSCT, and only 1 patient was alive after donor lymphocyte infusion (DLI). In total, of the 27

relapsed patients, 5 were still alive, 5 were lost to follow-up, and the remaining 17 died: 6

after second allo-HSCT, 8 after chemotherapy, and 3 after discontinuation of GVHD

prophylaxis. Thus, it is obvious that the outcomes of patients who experienced

hematological relapse after allo-HSCT were dismal. On the other hand, 13 patients who did

not experience hematological relapse but had MRD were all alive. Of these patients, 3 had

WT1 vaccination, 1 received second allo-HSCT for graft failure, and 9 experienced

13

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 14: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

spontaneous disappearance of MRD. The level of WT1 elevation in this group was only

between 100 and 200 copy/gRNA. The WT1 threshold to actively consider intervention to

prevent hematological relapse has not yet been established. Another important issue is the

continued lack of clarity regarding the most effective treatment strategy to prevent

hematological relapse at the time of MRD relapse after allo-HSCT. Possible preemptive

treatment interventions in patients with MRD relapse include azacitidine (Aza), FLT3

inhibitors, DLI, and second allo-HSCT as previously reported (ref. 25-31). Recently, Pozzi S

et al reported that preemptive treatment with DLI after allo-HSCT in patients with elevated

WT1 level improved prognosis (ref. 25). In their report, the risk of leukemia relapse was not

significantly reduced, although OS was significantly better in patients who received DLI

than in those who did not. These results may indicate that DLI alone is insufficient to

prevent hematological relapse. Regarding this point, Schroeder T et al showed that the

combination of Aza and DLI was an effective treatment strategy in patients with relapse

after allo-HSCT, in particular those with MDS or with AML characterized by low tumor

burden (ref. 28). In AML patients with high tumor burdens, the effects of these therapies

were limited. Therefore, it is important to detect MRD relapse using serial monitoring of

MRD after allo-HSCT. In addition to these reports, others showed that Aza after allo-HSCT

could induce expansion of immunomodulatory regulatory T cells and enhance the response

of cytotoxic T cells to tumor antigens (ref. 32, 33). Moreover, Aza could induce leukemic cell

differentiation and increase the expression of several tumor-associated antigens (ref. 32,

14

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 15: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

33). These effects of Aza after allo-HSCT may increase the graft-versus-leukemia effect

and improve patient outcomes (ref. 29-31). Prospective studies assessing the effectiveness

of preemptive treatment strategies incorporating such drugs are warranted.

There are several limitations to this study. First, we divided patients into three groups

based on their peripheral blood WT1 levels: WT1 <50 copy/gRNA, WT1 50-500

copy/gRNA, and WT1 >500 copy/gRNA. However, the cutoff value for peripheral WT1

level has not yet been well established. In our data-base, ROC curves at each time point to

determine the optimal cutoff values showed different describe each cutoff values: 600

copy/gRNA before allo-HSCT, 87 copy/gRNA at day 30, and 120 copy/gRNA at day 60

each other. However, it is practically complicated if we use different cutoff values at different

time points. Thus, we adopted more simplified cutoff values 50 and 500 copy/gRNA.

However, these values were arbitrary and the importance of these cutoffs should be

reconfirmed in other studies. The second limitation is that we did not use flow cytometry to

monitor MRD level routinely in clinical practice. Thus, to assess the benefit or limitation of

WT1 level in comparison to flow cytometry, further studies which incorporate both

measurements are needed. The third limitation is that this study consisted of a

retrospective analysis at a single center. Prospective studies are needed to establish

appropriate management approaches to elevated WT1 levels after allo-HSCT.

In conclusion, our data suggested that monitoring WT1 levels in PB after allo-HSCT

might be useful to identify patients at high risk of hematological relapse. Further

15

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 16: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

prospective studies are necessary to determine how to effectively and efficiently prevent

hematological relapse in patients diagnosed with MRD.

Acknowledgments

The authors thank the inpatient, outpatient, and support staff for their excellent care. This

work was supported by grants from the National Cancer Research and Development Fund

(26-A-26) and the Advanced Clinical Research Organization

Conflict of interest

Authors declare no conflict of interest.

References

1. Chen X, Xie H, Wood BL, et al. Relation of clinical response and minimal residual

disease and their prognostic impact on outcome in acute myeloid leukemia. J Clin Oncol.

2015;33:1258-1264.

2. Candoni A, Toffoletti E, Gallina R, et al. Monitoring of minimal residual disease by

quantitative WT1 gene expression following reduced intensity conditioning allogeneic stem

cell transplantation in acute myeloid leukemia. Clin Transplant. 2011;25:308-316.

3. Walter RB, Gooley TA, Wood BL, et al. Impact of pretransplantation minimal residual

disease, as detected by multiparametric flow cytometry, on outcome of myeloablative

16

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 17: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

hematopoietic cell transplantation for acute myeloid leukemia. J Clin Oncol. 2011;29:1190-

1197.

4. Ogawa H, Tamaki H, Ikegame K, et al. The usefulness of monitoring WT1 gene

transcripts for the prediction and management of relapse following allogeneic stem cell

transplantation in acute type leukemia. Blood. 2003;101:1698-1704.

5. Walter RB, Buckley SA, Pagel JM, et al. Significance of minimal residual disease before

myeloablative allogeneic hematopoietic cell transplantation for AML in first and second

complete remission. Blood. 2013;122:1813-1821.

6. Díez-Campelo M, Pérez-Simón JA, Pérez J, et al. Minimal residual disease monitoring

after allogeneic transplantation may help to individualize post-transplant therapeutic

strategies in acute myeloid malignancies. Am J Hematol. 2009;84:149-152.

7. Miyazaki T, Fujita H, Fujimaki K, et al. Clinical significance of minimal residual disease

detected by multidimensional flow cytometry: serial monitoring after allogeneic stem cell

transplantation for acute leukemia. Leuk Res. 2012;36: 998-1003.

8. Campana D, Leung W. Clinical significance of minimal residual disease in patients with

acute leukaemia undergoing haematopoietic stem cell transplantation. Br J Haematol.

2013;162:147-161.

9. Jacobsohn DA, Tse WT, Chaleff S, et al. High WT1 gene expression before

haematopoietic stem cell transplant in children with acute myeloid leukaemia predicts poor

event-free survival. Br J Haematol. 2009;146:669-674.

17

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 18: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

10. Call KM, Glaser T, Ito CY, et al. Isolation and characterization of a zinc finger

polypeptide gene at the human chromosome 11 Wilms' tumor locus. Cell. 1990;60:509-520.

11. Gessler M, Poustka A, Cavenee W, et al. Homozygous deletion in Wilms tumours of a

zinc-finger gene identified by chromosome jumping. Nature. 1990;343:774-778.

12. Inoue K, Ogawa H, Sonoda Y, et al. Aberrant overexpression of the Wilms tumor gene

(WT1) in human leukemia. Blood. 1997;89:1405-1412.

13. Inoue K, Sugiyama H, Ogawa H, et al. WT1 as a new prognostic factor and a new

marker for the detection of minimal residual disease in acute leukemia. Blood.

1994;84:3071-3079.

14. Barragán E, Cervera J, Bolufer P, et al. Prognostic implications of Wilms' tumor gene

(WT1) expression in patients with de novo acute myeloid leukemia. Haematologica.

2004;89:926-933.

15. Cilloni D, Gottardi E, Messa F, et al. Significant correlation between the degree of WT1

expression and the International Prognostic Scoring System Score in patients with

myelodysplastic syndromes. J Clin Oncol. 2003;21:1988-1995.

16. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain

reaction detection of minimal residual disease by standardized WT1 assay to enhance risk

stratification in acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol.

2009;27:5195-5201.

17. Yamauchi T, Negoro E, Lee S, et al. Detectable Wilms' tumor-1 transcription at

18

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 19: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

treatment completion is associated with poor prognosis of acute myeloid leukemia: a single

institution's experience. Anticancer Res. 2013;33:3335-3340.

18. Gianfaldoni G, Mannelli F, Ponziani V, et al. Early reduction of WT1 transcripts during

induction chemotherapy predicts for longer disease free and overall survival in acute

myeloid leukemia. Haematologica. 2010;95:833-836.

19. Gray JX, McMillen L, Mollee P, et al. WT1 expression as a marker of minimal residual

disease predicts outcome in acute myeloid leukemia when measured post-consolidation.

Leuk Res. 2012;36:453-458.

20. Lapillonne H, Renneville A, Auvrignon A, et al. High WT1 expression after induction

therapy predicts high risk of relapse and death in pediatric acute myeloid leukemia. J Clin

Oncol. 2006;24:1507-1515.

21. Weisser M, Kern W, Rauhut S, et al. Prognostic impact of RT-PCR-based quantification

of WT1 gene expression during MRD monitoring of acute myeloid leukemia. Leukemia.

2005;19:1416-1423.

22. Kitamura K, Nishiyama T, Ishiyama K, et al. Clinical usefulness of WT1 mRNA

expression in bone marrow detected by a new WT1 mRNA assay kit for monitoring acute

myeloid leukemia: a comparison with expression of WT1 mRNA in peripheral blood. Int J

Hematol. 2016;103:53-62.

23. Kanda Y. Investigation of the freely available easy-to-use software 'EZR' for medical

statistics. Bone Marrow Transplant. 2013;48:452-458.

19

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 20: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

24. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating

prognosis in myelodysplastic syndromes. Blood. 1997;89:2079-2088.

25. Pozzi S, Geroldi S, Tedone E, et al. Leukaemia relapse after allogeneic transplants for

acute myeloid leukaemia: predictive role of WT1 expression. Br J Haematol. 2013;160:503-

509.

26. Sammons SL, Pratz KW, Smith BD, Karp JE, Emadi A. Sorafenib is tolerable and

improves clinical outcomes in patients with FLT3-ITD acute myeloid leukemia prior to stem

cell transplant and after relapse post-transplant. Am J Hematol. 2014;89:936-938.

27. Metzelder SK, Schroeder T, Finck A, et al. High activity of sorafenib in FLT3-ITD-

positive acute myeloid leukemia synergizes with allo-immune effects to induce sustained

responses. Leukemia. 2012;26:2353-2359.

28. Schroeder T, Rachlis E, Bug G, et al. Treatment of acute myeloid leukemia or

myelodysplastic syndrome relapse after allogeneic stem cell transplantation with azacitidine

and donor lymphocyte infusions--a retrospective multicenter analysis from the German

Cooperative Transplant Study Group. Biol Blood Marrow Transplant. 2015;21:653-660.

29. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treatment of imminent relapse

in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia.

2012;26:381-389.

30. Craddock C, Jilani N, Siddique S, et al. Tolerability and Clinical Activity of Post-

Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on

20

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 21: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

the RICAZA Trial. Biol Blood Marrow Transplant. 2016;22:385-390.

31. Steinmann J, Bertz H, Wäsch R, et al. 5-Azacytidine and DLI can induce long-term

remissions in AML patients relapsed after allograft. Bone Marrow Transplant. 2015;50:690-

695.

32. Choi J, Ritchey J, Prior JL, et al. In vivo administration of hypomethylating agents

mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. Blood.

2010;116:129-139.

33. Goodyear OC, Dennis M, Jilani NY, et al. Azacitidine augments expansion of regulatory

T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia

(AML). Blood. 2012;119:3361-3369.

21

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 22: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Table 1. Clinical characteristics of study population (N=98)Characteristic No. %No. of patients 98 100.0Age, years     Median 46.5   Range 18 to 68  Sex     Male 55 56.1 Female 43 43.9Disease     AML, MDS/AML 84 85.7 MDS 14 14.3Status of disease     CR 49 50.0 Non-CR 49 50.0Secondary AML     No 90 91.8 Yes 8 8.2Cytogenetic risk group     Good 47 47.9 Intermediate 27 27.6 Poor 20 20.4 Not available 4 4.1Relation to donor     Related donor 27 27.5 Unrelated donor 71 72.5Stem cell source     Bone marrow 65 66.3 Peripheral blood 27 27.6 Cord blood 6 6.1HLA compatibility     Fully matched 45 45.9 1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6Conditioning regimen     TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5GVHD prophylaxis     Tac-based 78 79.6 CyA-based 17 17.4 Post CY 3 3.0ATG     No 84 85.7

Yes 14 14.3Abbreviations: AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; CR, complete remission; HLA, human leukocyte antigen; TBI, total body irradiation; CY, cyclophosphamide; Bu, busulfan; Flu, fludarabine; GVHD, graft-versus-host-disease; Tac,

22

1

2

345

Page 23: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

tacrolimus; CyA, cyclosporine A; ATG, anti-thymocyte globulin.

Table 2. Univariate analysis of CIRFactor HR 95% CI PDisease AML, MDS/AML (n=84) 1.00 MDS (n=14) 1.06 0.42 to 2.69 0.89Status of disease       CR (n=49) 1.00     Non-CR (n=49) 5.16 2.46 to 10.82 <0.01Sex       Male (n=55) 1.00 Female (n=43) 0.40 0.21 to 0.78 <0.01Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.48 0.68 to 3.23 0.32 Poor (n=20) 2.89 1.53 to 5.49 <0.01Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 1.43 0.72 to 2.86 0.31WT1 before allo-HSCT       <50 (n=34) 1.00     50-500 (n=13) 1.40 0.35 to 5.58 0.63 >500 (n=41) 4.22 2.04 to 8.76 <0.01WT1 at day 30 after allo-HSCT     <50 (n=78) 1.00 50-500 (n=13) 1.60 0.57 to 4.47 0.37 >500 (n=6) 7.43 2.17 to 25.43 <0.01WT1 at day 60 after allo-HSCT     <50 (n=77) 1.00     50-500 (n=8) 2.75 0.89 to 8.43 0.08 >500 (n=5) 24.73 8.32 to 73.51 <0.01

Abbreviations: CIR, cumulative incidence of relapse; HR, hazard ratio; CI, confidence interval; allo-HSCT, allogeneic hematopoietic stem cell transplantation.

23

123

45

6

7

8

9

10

11

Page 24: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Table 3. Multivariate analysis of CIR: before and at days 30 and 60 after allo-HSCTFactor HR 95% CI P

Before allo-HSCTDisease AML, MDS/AML (n=75) 1.00 MDS (n=13) 0.25 0.09 to 0.70 <0.01Status of disease       CR (n=44) 1.00     Non-CR (n=44) 4.40 1.56 to 12.42 <0.01Sex       Male (n=50) 1.00 Female (n=38) 0.26 0.13 to 0.53 <0.01Cytogenetic risk group       Good (n=41) 1.00     Intermediate (n=25) 2.35 0.71 to 7.83 0.16 Poor (n=19) 1.57 0.71 to 3.47 0.27Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 3.03 0.79 to 11.69 0.11WT1 before allo-HSCT       <50 (n=34) 1.00     50-500 (n=13) 0.93 0.15 to 5.93 0.94 >500 (n=41) 3.00 1.20 to 7.49 0.02

Day 30 after allo-HSCTDisease AML, MDS/AML (n=83) 1.00 MDS (n=14) 0.58 0.28 to 1.20 0.15Status of disease       CR (n=49) 1.00     Non-CR (n=48) 5.04 2.25 to 11.27 <0.01Sex       Male (n=54) 1.00 Female (n=43) 0.29 0.13 to 0.65 <0.01Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.73 0.73 to 4.11 0.22 Poor (n=19) 1.79 0.77 to 4.19 0.18Conditioning regimen MAC (n=71) 1.00 RIC (n=26) 1.58 0.81 to 3.10 0.18WT1 at day 30 after allo-HSCT     <50 (n=78) 1.00     50-500 (n=13) 2.98 1.19 to 7.47 0.02 >500 (n=6) 7.86 2.28 to 27.12 <0.01

Day 60 after allo-HSCTDisease AML, MDS/AML (n=77) 1.00 MDS (n=13) 0.23 0.05 to 1.05 0.06Status of disease       CR (n=49) 1.00     Non CR (n=41) 3.88 1.77 to 8.48 <0.01Sex       Male (n=49) 1.00 Female (n=41) 0.54 0.27 to 1.09 0.09Cytogenetic risk group       Good (n=45) 1.00     Intermediate (n=25) 1.61 0.59 to 4.35 0.35 Poor (n=16) 1.41 0.64 to 3.11 0.39

24

1

2

Page 25: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Conditioning regimen MAC (n=60) 1.00 RIC (n=20) 2.39 0.45 to 12.64 0.31WT1 at day 60 after allo-HSCT     <50 (n=77) 1.00     50-500 (n=8) 2.94 0.88 to 9.82 0.08 >500 (n=5) 15.90 4.22 to 59.92 <0.01

Table 4. Univariate analysis of OSFactor HR 95% CI PDisease AML, MDS/AML (n=84) 1.00 MDS (n=14) 0.59 0.14 to 2.51 0.47Status of disease       CR (n=49) 1.00     Non-CR (n=49) 6.08 2.28 to 16.21 <0.01Sex       Male (n=55) 1.00 Female (n=43) 0.58 0.26 to 1.31 0.19Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.03 0.38 to 2.77 0.96 Poor (n=20) 3.08 1.35 to 7.02 <0.01Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 1.38 0.60 to 3.18 0.45WT1 before allo-HSCT       <50 (n=34) 1.00     50-500 (n=13) 1.68 0.31 to 9.23 0.55 >500 (n=41) 5.41 2.13 to 13.76 <0.01WT1 at day 30 after allo-HSCT     <50 (n=78) 1.00 50-500 (n=13) 0.78 0.18 to 3.34 0.73 >500 (n=7) 5.11 1.70 to 15.34 <0.01WT1 at day 60 after allo-HSCT     <50 (n=77) 1.00     50-500 (n=9) 3.20 1.04 to 9.87 0.04 >500 (n=8) 25.78 6.62 to 100.40 <0.01

25

1

2

3

4

5

Page 26: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Table 5. Multivariate analysis of OS: before and at days 30 and 60 after allo-HSCTFactor HR 95% CI P

Before allo-HSCTDisease AML, MDS/AML (n=75) 1.00 MDS (n=13) 0.12 0.02 to 0.88 0.04Status of disease       CR (n=44) 1.00     Non-CR (n=44) 9.00 3.32 to 24.44 <0.01Sex       Male (n=50) 1.00 Female (n=38) 0.43 0.18 to 1.00 0.05Cytogenetic risk group       Good (n=41) 1.00     Intermediate (n=25) 1.78 0.55 to 5.70 0.33 Poor (n=19) 1.57 0.63 to 3.94 0.34Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 2.29 0.44 to 12.06 0.33WT1 before allo-HSCT       <50 (n=34) 1.00     50-500 (n=13) 1.42 0.21 to 9.58 0.72 >500 (n=41) 2.55 0.78 to 8.29 0.12

Day 30 after allo-HSCTDisease AML, MDS/AML (n=84) 1.00 MDS (n=14) 0.29 0.07 to 1.25 0.09Status of disease       CR (n=49) 1.00     Non-CR (n=49) 6.09 2.29 to 16.22 <0.01Sex       Male (n=55) 1.00 Female (n=43) 0.58 0.26 to 1.33 0.20Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.65 0.57 to 4.76 0.35 Poor (n=20) 2.33 0.99 to 5.48 0.05Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 1.60 0.66 to 3.89 0.30WT1 at day 30 after allo-HSCT     <50 (n=78) 1.00     50-500 (n=13) 1.29 0.25 to 6.67 0.76 >500 (n=7) 2.29 0.56 to 9.38 0.25

Day 60 after allo-HSCTDisease AML, MDS/AML (n=80) 1.00 MDS (n=14) 0.25 0.05 to 1.12 0.07Status of disease      

26

1

2

3

4

5

Page 27: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

CR (n=49) 1.00     Non-CR (n=45) 4.32 1.55 to 12.04 <0.01Sex       Male (n=53) 1.00 Female (n=41) 0.56 0.21 to 1.53 0.26Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.26 0.36 to 4.43 0.72 Poor (n=18) 1.39 0.43 to 4.54 0.58Conditioning regimen MAC (n=70) 1.00 RIC (n=24) 2.64 0.44 to 15.86 0.29WT1 at day 60 after allo-HSCT     <50 (n=77) 1.00     50-500 (n=9) 2.90 0.94 to 8.93 0.06 >500 (n=8) 13.94 3.59 to 54.11 <0.01

Figure legends

Figure 1. Monitoring and change of peripheral blood WT1 levels before and after allo-

HSCT.

Data indicate the WT1 levels before and 30 and 60 days after allo-HSCT. The upper row of

text in each box indicates patient group based on WT1 levels (units: copy/gRNA), and the

lower row indicates the number of patients corresponding to each box.

Abbreviations: allo-HSCT, allogeneic hematopoietic stem cell transplantation; NA, not

available.

Figure 2. Cumulative incidence of relapse and overall survival before and after allo-

HSCT according to MRD levels.

Solid line, WT1 <50 copy/gRNA; broken line, WT1 50-500 copy/gRNA; dotted line, WT1

>500 copy/gRNA. a, b; before allo-HSCT, c, d; day 30 after allo-HSCT, e, f; day 60 after

27

1

2

3

4

5

6

7

8

9

10

11

12

13

14

Page 28: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

allo-HSCT. Abbreviations: CI, confidence interval.

Figure 3. The prognosis of cases with elevated WT1.

Of the 27 patients with hematological relapse, only 5 were alive at the end of the study. In

contrast, all 13 patients with MRD but no hematological relapse survived.

Abbreviations: SCT, stem cell transplantation; DLI, donor lymphocyte infusion; GVL, graft

versus leukemia.

28

1

2

3

4

5

6

7

Page 29: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Figure 1.

29

1

2

Page 30: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Figure 2.

30

1

2

Page 31: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Figure 3.

31

1

2

3

4

5

Page 32: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Table 1. Transition of WT1 in PB based on MAC or RIC conditioning

Data of WT1 in PB day 0 (N=72) day 30 (N=72) day 60 (N=70)

MAC N % N % N %<50 27 37.5 58 80.5 59 84.3

50-500 12 16.7 11 15.3 7 10.0>500 26 36.1 3 4.2 4 5.7NA 7 9.7 0 0.0 0 0.0

RIC day 0 (N=26) day 30 (N=26) day 60 (N=24)

<50 7 26.9 20 76.9 18 75.050-500 1 3.9 2 7.7 2 8.3>500 15 57.7 4 15.4 4 16.7NA 3 11.5 0 0.0 0 0.0

With regard to the conditioning intensity, a univariate analysis showed a trend toward

higher rate of proportion of MRD negativity in patients who received a myeloablative

conditioning regimen.

Supplemental Table 2. WT1 data at day 90, 180 of allo-HSCT

  Day 90 of allo-HSCT (N=85) Day 180 of allo-HSCT(N=76)

Data of WT1 in PB Total No. of relapse No. of relapse / Total Total No. of relapse No. of relapse / Total

  N N %     %

<50 74 19 26.7 69 11 15.9

50-500 7 4 57.1 2 2 100

>500 2 2 100 3 3 100

NA 2 2 100 2 1 50

At day 90, in 9 patients of MRD positivity, 6 patients had subsequent hematological relapse,

and at day 180, in 5 patients of MRD positivity, all had hematological relapse.

32

1

2

3

4

5

6

7

8

9

10

11

12

Page 33: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure legends

Supplemental Figure 1. Cumulative incidence of relapse and overall survival before

and after allo-HSCT in hematological CR patients (N=49).

In hematological CR patients, WT1 level of PB was relatively related to CIR. On the other

hand, WT1 level of PB was not related to OS.

Solid line, WT1 <50 copy/gRNA; broken line, WT1 50-500 copy/gRNA; dotted line, WT1

>500 copy/gRNA.

a, b; before allo-HSCT, c, d; day 30 after allo-HSCT, e, f; day 60 after allo-HSCT.

Abbreviations: allo-HSCT, allogeneic hematopoietic stem cell transplantation; CI,

confidence interval; NA, not available.

Supplemental Figure 2. Cumulative incidence of relapse and overall survival before

and after allo-HSCT in hematological non-CR patients (N=49).

In hematological non-CR patients, WT1 level of PB was relatively related to CIR and OS at

the time of day 30 and 60.

Supplemental Figure 3. Cumulative incidence of relapse and overall survival before

and after allo-HSCT in AML patients (N=84).

In AML patients, WT1 level of PB was significantly related to CIR and OS.

33

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 34: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure 4. Cumulative incidence of relapse and overall survival before

and after allo-HSCT in MDS patients (N=14).

In MDS patients, the number of patients was small, the analysis was not detectable.

Supplemental Figure 5. Cumulative incidence of relapse and overall survival before

and after allo-HSCT according to MRD levels (Cutoff value of WT1 200 copy/gRNA).

In this analysis, WT1 level was divided into 3 groups according to <50 copy/gRNA, 50-200

copy/gRNA and >200 copy/gRNA. The cutoff value was different from the analysis of

above. CIR and OS differed statistically (P<0.05) according to the WT1 levels before and at

day 30 and 60 after allo-HSCT.

Solid line, WT1 <50 copy/gRNA; broken line, WT1 50-200 copy/gRNA; dotted line, WT1

>200 copy/gRNA.

a, b; before allo-HSCT, c, d; day 30 after allo-HSCT, e, f; day 60 after allo-HSCT.

Abbreviations: CI, confidence interval.

34

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

Page 35: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure 1. CIR and OS before and after allo-HSCT according to MRD

levels in hematological CR patients (N=49).

35

1

2

3

Page 36: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure 2. CIR and OS before and after allo-HSCT according to MRD

levels in hematological non-CR patients (N=49).

36

1

2

3

Page 37: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure 3. CIR and OS before and after allo-HSCT according to MRD

levels in AML patients (N=84).

37

1

2

3

Page 38: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure 4. CIR and OS before and after allo-HSCT according to MRD

levels in MDS patients (N=14).

38

1

2

3

Page 39: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Figure 5. CIR and OS before and after allo-HSCT according to MRD

levels (WT1 cutoff value 200 copy/gRNA).

39

1

2

3

Page 40: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Supplemental Table 3. Multivariate analysis of CIR: before and at days 30 and 60 after allo-HSCT (WT1 cutoff value 200 copy/gRNA)Factor HR 95% CI P

Before allo-HSCTDisease AML, MDS/AML (n=75) 1.00 MDS (n=13) 0.31 0.11 to 0.85 0.02Status of disease       CR (n=44) 1.00     Non-CR (n=44) 7.91 3.32 to 18.79 <0.01Sex       Male (n=50) 1.00 Female (n=38) 0.33 0.17 to 0.65 <0.01Cytogenetic risk group       Good (n=41) 1.00     Intermediate (n=25) 1.68 0.64 to 4.45 0.29 Poor (n=19) 1.86 0.83 to 4.15 0.13Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 2.91 0.78 to 10.78 0.11WT1 before allo-HSCT       <50 (n=34) 1.00     50-200 (n=9) 1.45 0.24 to 8.90 0.69 >200 (n=45) 1.71 0.68 to 4.31 0.25

Day 30 after allo-HSCTDisease AML, MDS/AML (n=83) 1.00 MDS (n=14) 0.54 0.22 to 1.33 0.18Status of disease       CR (n=49) 1.00     Non-CR (n=48) 5.02 2.17 to 11.60 <0.01Sex       Male (n=54) 1.00 Female (n=43) 0.29 0.12 to 0.66 <0.01Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.69 0.80 to 3.61 0.17 Poor (n=19) 1.85 0.77 to 4.45 0.17Conditioning regimen MAC (n=71) 1.00 RIC (n=26) 1.56 0.76 to 3.22 0.22WT1 at day 30 after allo-HSCT     <50 (n=78) 1.00     50-200 (n=11) 3.02 1.15 to 7.96 0.03 >200 (n=8) 6.32 2.05 to 19.42 <0.01

Day 60 after allo-HSCTDisease AML, MDS/AML (n=77) 1.00 MDS (n=13) 0.39 0.16 to 0.92 0.03Status of disease       CR (n=49) 1.00     Non CR (n=41) 5.82 2.26 to 14.97 <0.01Sex       Male (n=49) 1.00 Female (n=41) 0.38 0.17 to 0.86 0.02Cytogenetic risk group       Good (n=45) 1.00     Intermediate (n=25) 2.12 0.82 to 5.52 0.12 Poor (n=16) 1.66 0.78 to 3.51 0.19Conditioning regimen MAC (n=60) 1.00

40

12

Page 41: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

RIC (n=20) 1.47 0.71 to 3.04 0.30WT1 at day 60 after allo-HSCT     <50 (n=77) 1.00     50-200 (n=5) 4.86 1.38 to 17.15 0.01 >200 (n=8) 5.62 1.84 to 17.16 <0.01

Supplemental Table 4. Multivariate analysis of OS: before and at days 30 and 60 after allo-HSCT (WT1 cutoff value 200 copy/gRNA)Factor HR 95% CI P

Before allo-HSCTDisease AML, MDS/AML (n=75) 1.00 MDS (n=13) 0.12 0.02 to 0.88 0.04Status of disease       CR (n=44) 1.00     Non-CR (n=44) 9.00 3.32 to 24.44 <0.01Sex       Male (n=50) 1.00 Female (n=38) 0.53 0.23 to 1.22 0.14Cytogenetic risk group       Good (n=41) 1.00     Intermediate (n=25) 1.75 0.53 to 5.76 0.36 Poor (n=19) 1.88 0.73 to 4.83 0.19Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 3.07 0.60 to 15.71 0.18WT1 before allo-HSCT       <50 (n=34) 1.00     50-200 (n=9) 1.58 0.15 to 16.21 0.70 >200 (n=45) 2.60 0.82 to 8.19 0.10

Day 30 after allo-HSCTDisease AML, MDS/AML (n=84) 1.00 MDS (n=14) 0.29 0.07 to 1.25 0.09Status of disease       CR (n=49) 1.00     Non-CR (n=49) 6.09 2.29 to 16.22 <0.01Sex       Male (n=55) 1.00 Female (n=43) 0.58 0.26 to 1.33 0.20Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.68 0.58 to 4.84 0.34 Poor (n=20) 2.33 0.99 to 5.48 0.05Conditioning regimen MAC (n=72) 1.00 RIC (n=26) 1.60 0.66 to 3.89 0.30WT1 at day 30 after allo-HSCT     <50 (n=78) 1.00     50-200 (n=11) 1.70 0.32 to 9.10 0.54 >200 (n=9) 1.86 0.49 to 7.10 0.36

Day 60 after allo-HSCTDisease AML, MDS/AML (n=80) 1.00 MDS (n=14) 0.25 0.05 to 1.22 0.08Status of disease       CR (n=49) 1.00     Non-CR (n=45) 4.48 1.62 to 12.39 <0.01Sex       Male (n=53) 1.00 Female (n=41) 0.53 0.20 to 1.40 0.20

41

123

Page 42: Kazuko Ino · Web view1 locus mismatched 23 23.5 >2-loci mismatched 30 30.6 Conditioning regimen TBI/CY 28 28.6 Bu/CY 32 32.7 Flu/Bu4 12 12.2 Flu/Bu2 26 26.5 GVHD prophylaxis Tac-based

Cytogenetic risk group       Good (n=47) 1.00     Intermediate (n=27) 1.69 0.51 to 5.55 0.39 Poor (n=18) 1.69 0.45 to 6.45 0.44Conditioning regimen MAC (n=70) 1.00 RIC (n=24) 3.74 0.68 to 20.42 0.13WT1 at day 60 after allo-HSCT     <50 (n=77) 1.00     50-200 (n=5) 4.17 0.88 to 19.81 0.07 >200 (n=12) 6.09 2.36 to 15.72 <0.01

42

1