Zum Inhalt

Impact of BCOR/BCORL1 mutation on outcomes of allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia patients

  • Open Access
  • 09.04.2025
  • Review
Erschienen in:
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

BCOR alteration is a well-established adverse-risk marker for acute myeloid leukemia (AML) in 2022 ELN risk stratification. However, outcomes of BCOR- or BCORL1-mutated AML after allogeneic hematopoietic stem cell transplantation (allo-HSCT) are as yet poorly defined. In an 877-patient consecutive AML transplantation cohort, we found 83 (9.5%) patients with BCOR or BCORL1 mutation (BCOR/BCORL1mut). We retrospectively evaluated the clinical characteristics and transplant outcomes of BCOR/BCORL1mut patients and compared them with 276 patients with normal karyotype (BCOR/BCORL1wt). Frameshift mutation was the predominant alteration of BCOR (n = 22, 39.3%), and the majority of BCORL1 was missense mutation (n = 25, 65.8%). The most common co-mutated gene of BCOR/BCORL1mut was DNMT3A (n = 23, 27.7%). BCOR/BCORL1mut was also associated with lower WBC counts at diagnosis (P = 0.003), shorter interval from diagnosis to transplantation (P = 0.037), and fewer achieved minimal residual disease negativity pre-transplantation (P < 0.001), compared to BCOR/BCORL1wt. Three-year OS, DFS and CIR of BCOR/BCORL1wt and BCOR/BCORL1mut groups were 75.2% (95% CI, 70.0-80.8%) vs. 76.0% (95% CI, 66.0-87.5%) (HR, 0.92; 95% CI, 0.54–1.57; P = 0.77), 74.5% (95% CI, 69.4-80.1%) vs. 67.7% (95%CI, 57.0-80.4%) (HR, 1.20; 95% CI, 0.75–1.91; P = 0.46), and 12.6% (95% CI, 8.9-17.0%) vs. 24.0% (95% CI, 14.1-35.4%) (HR, 1.85; 95% CI, 1.04–3.3; P = 0.03), respectively. We also investigated the impact of the type and location of BCOR/BCORL1mut on transplant outcomes, but no significant effect was observed. Our findings suggest that BCOR/BCORL1mut is associated with relapse after allo-HSCT, despite no observed difference in OS, and that allo-HSCT could help to overcome the impact of BCOR/BCORL1mut characteristics on outcomes.

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00277-025-06346-6.
YunXia Zhou and Haixiao Zhang these authors contributed equally.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

The gene BCOR, a transcription factor, located on chromosome X at p11.4, is involved in lymphocyte development, function of hematopoietic and mesenchymal stem cells, and pluripotency of embryonic stem cells [1]. It was first identified in a 2-hybrid screen for interactors with transcriptional repressor BCL6, whose alteration can lead to diffuse large B cell lymphomas [2]. Genetic lesions of BCOR have also been shown to be associated with hematopoietic malignancies [3]. Researchers have detected BCOR mutations were detected in 3.8–5.0% of adult de novo acute myeloid leukemia (AML) and 8% of secondary AML [46], previous studies have confirmed that BCOR mutations are associated with adverse prognosis in AML. As a result, they were classified into the adverse-risk group as per the 2022 European Leukemia Net (ELN) recommendations [4, 711]. Furthermore, BCOR mutations have also been found to be involved in the proliferation and differentiation of hematopoietic cells and to play an important role in transcriptional and epigenetic regulation [4, 12]. They also cause disorderly proliferation of myeloid progenitor cells and inhibit differentiation [1, 12, 13].
BCORL1, located at chromosome Xq26.1, is a homologous gene of BCOR. The prevalence of BCORL1 mutations is similar to BCOR-mutated AML, at 3.7–5.8% in AML adult patients [5, 14], but despite their many similarities, the BCOR and BCORL1 genes exhibit some different features in terms of the capability to interact with BCL6, localization within subnuclear structures, and expression levels in human tissues [6].
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only potentially curative treatment for many high-risk AML patients [11, 15, 16]. However, although the impact of BCOR and/or BCORL1 mutations (BCOR/BCORL1mut) on AML patients are well-established in chemotherapy cohorts, the clinical significance of BCOR/BCORL1mut is less clear in transplant patients. We therefore performed a retrospective analysis in an attempt to determine the clinical characteristics and transplant outcomes of BCOR- and BCORL1-mutated AML.

Subjects and methods

Subjects enrollment and definitions

We retrospectively investigated a cohort of 877 consecutive adult AML subjects, who received their first allo-HSCT from January, 2018 to December, 2023, based on the National Longitudinal Cohort of Hematological Diseases (NICHE, NCT04645199) from the Institute of Hematology and Blood Diseases Hospital, Peking Union Medical College, and the Chinese Academy of Medical Sciences (IHCAMS). Eventually, 83 (9.5%) subjects with BCOR/BCORL1mut at diagnosis were identified. Additionally, we also enrolled 276 AML patients with normal karyotypes, defined as the BCOR/BCORL1wt group (Fig. 1a).
Fig. 1
Subjects enrollment, and characteristic of BCOR and BCORL1 mutation. (a) Criteria for inclusion and exclusion of subjects. (b) Lollipop plot showing various mutations across the BCOR and BCORL1 mutations. (c) Co-mutational profile in BCOR/BCORL1-mutated and normal karyotype AML. (d) 2022 ELN risk stratification and variant allele frequency (VAF) in BCOR/BCORL1-mutated patients
Bild vergrößern
The following criteria were utilized to diagnose and evaluate patient disease status. The diagnosis of AML adhered to the 2016 World Health Organization (WHO) guidelines and disease risk was assessed using the 2022 ELN [11, 17]. Minimal residual disease negativity (MRD) was assessed using multiparameter flow cytometry, and MRD positivity was defined as levels > 0.01%. The definition of complete remission (CR) included bone marrow (BM) blasts of less than 5%, the absence of circulating blasts, and no extramedullary disease [11]. Relapse was defined as the presence of 5% or more leukemic blasts in the BM, the reappearance of leukemic blasts in peripheral blood (PB) in at least two PB samples taken at least one week apart, or the emergence of new extramedullary disease [11]. Patients were classified as having refractory disease if a specified response was not achieved by a designated landmark (such as failure to attain a response after two cycles of intensive chemotherapy) or a predetermined landmark (for example, 180 days after initiating less-intensive therapy) [11].
This study was approved by the Ethics Committee at IHCAMS and adhered to the Declaration of Helsinki. All patients provided written informed consent to participate.

Targeted gene sequencing

The protocol for carrying out targeted gene sequencing has been described previously [18]. In brief, genomic DNA was extracted from BM specimens, targeting at least 141 genes involved in point mutations and insertion/deletion mutations in protein-coding regions or hotspots (Table S1). Exons of these genes were sequenced with a depth of 800×, ion and variant reporters were used for mutation analysis. Databases such as dbSNP, 1000 Genomes, Polyphen-2, and COSMIC were used for mutation annotation and amino acid mutation analysis. Using this method, the detection rate of mutation sites with 5% mutation frequency was more than 97%.

Transplantation procedure

All patients included in this study received intensive induction chemotherapy prior to allo-HSCT. The standard anthracycline and cytarabine-based regimen was used for induction chemotherapy, followed by consolidation chemotherapy, which typically involves primary induction chemotherapy or high-dose cytarabine. All patients received a modified myeloablative conditioning (MAC) regimens based on busulfan (Bu) and cyclophosphamide (Cy). For mismatched related donors and unrelated donors (URD), anti-thymocyte globulin (ATG, 2-2.5 mg/kg/day on days − 4 to -1) was required, and donor peripheral blood stem cells were collected following G-CSF mobilization (5–10 µg·kg− 1·day− 1 for 5–6 days). Cyclosporine (CAS) or Tacrolimus (FK506) in combination with a short course of methotrexate (MTX) with or without mycophenolate mofetil (MMF) were used for the prophylaxis of acute graft versus host donor (GVHD). Acute GVHD was graded according to MAGIC criteria and severity of chronic GVHD was determined using the 2014 National Institute of Health consensus criteria [19, 20]. Methylprednisolone was used as the first-line treatment for acute GVHD. Neutrophil engraftment was defined as the first neutrophil count more than 0.5 × 109 /L for 3 consecutive days and platelet engraftment was defined as the first platelet count more than 20 × 109 /L without transfusion for 7 consecutive days. The post-transplant disease status of BM was assessed at days + 14, +28, + 42 and months + 2, +3, + 4.5, + 6, +9 and + 12.

Statistical analysis

The statistical comparison of categorical variables was performed using chi-squared tests or Fisher’s exact tests and continuous variables were compared using t tests or Kruskal-Wallis tests. Overall survival (OS) and disease-free survival (DFS) were estimated using the Kaplan-Meier method and log-rank test. Cumulative incidence of relapse (CIR) and non-relapse mortality (NRM) were estimated using competing risk model analysis and Fine-Grey’s test. OS and DFS were defined as the time from transplantation to death and relapse or death, respectively; CIR was defined as the time from transplantation to relapse and with non-relapse mortality (NRM) as the time from transplantation to death without relapse. Univariate and multivariate analysis were both used to assess the association between survival outcomes and clinical characteristics of the patients. Variables with P values < 0.1 in univariate analysis were included in the multivariate Cox models and competing risk models. A P value < 0.05 was considered to indicate statistical significance in each of these multivariate models. Statistical analysis was performed using R 4.3.3 and SPSS 26.

Results

The BCOR/BCORL1mut landscape

Out of the 877 consecutive NGS tests performed, 83 (9.5%) subjects with BCOR/BCORL1mut were identified. BCOR and BCORL1 alterations occurred in 56 (6.4%) and 38 (4.3%) patients, respectively, with 11 (13.3%) instances of BCOR and BCORL1 co-mutation. BCOR mutations were frequent in exon 4 (43%), and BCORL1 mutations were frequent in exon 3 (77%) (Fig. 1c, d). Frameshift mutation was the predominant alteration of BCOR (n = 22, 39.3%), followed by missense variants (n = 18, 32.1%), and missense mutation account for the majority of BCORL1 mutations (n = 25, 65.8%). The plurality of alterations occurred outside the functional domains (n = 74, 89.2%). Alteration of AF9BD domain of BCOR, which directly interacts with the transcriptional regulator AF9 (MLLT3) [2, 21, 22], was present in 12.5% (n = 7) patients, and the ANK domain of BCORL1 was altered in 2 (5.3%) patients (Fig. 1b). The average variant allele frequency (VAF) of BCOR and BCORL1 genes were 42.7% (range 1.7-100.0%) and 48.1% (range 2.5-100.0%), respectively.
The median co-occurring mutations of BCOR/BCORL1mut was 4 (range 0–10), and the most common co-mutated gene was DNMT3A (n = 23, 27.7%), followed by genes NRAS (n = 21, 25.3%), FLT3 (n = 21, 25.3%), RUNX1 (n = 20, 24.1%), and WT1 (n = 14, 16.9%), but only 4 (4.8%) patients co-occured TP53 mutations. For FLT3-mutated patients, 13 had FLT3-ITD mutations and 8 had FLT3-TKD mutations, respectively. In BCOR/BCORL1wt patients, FLT3 (n = 114, 41.3%), NPM1 (n = 86, 31.2%), DNMT3A (n = 69, 25.0%), and CEBPA (n = 61, 22.1%) mutations were more frequent (Fig. 1c).
According to ELN 2022, 9 (10.8%) patients were classified as favorable-risk, including 3 with t(8;21), 3 with NPM1 mutation without FLT3-ITD mutation, and 3 with bZIP in-frame mutated CEBPA, 19 (22.9%) were intermediate-risk, and 55 (66.3%) were adverse-risk. Notably, the intermediate-risk patients had higher VAF compared to the adverse-risk patients (Median: 67.6 vs. 42.7, P < 0.01) (Fig. 1d). In terms of cytogenetic abnormality, 86.75% (n = 72) of BCOR/BCORL1mut patients had normal karyotypes.

Characteristics of BCOR/BCORL1mut

A total of 359 AML patients who received their first allo-HSCT were included in this study. The median age was 38 [interquartile range (IQR), 30–48] years. Of the 359 patients, 83 (23.1%) cases had BCOR/BCORL1mut and 276 (76.9%) patients with normal karyotypes were enrolled in the BCOR/BCORL1wt group. The clinical characteristics of the BCOR/BCORL1mut and BCOR/BCORL1wt groups are summarized in Table 1. The median age of the BCOR/BCORL1mut group was 38 (IQR, 33–48) years, similar to that of the BCOR/BCORL1wt group (38 [IQR, 29–49] years, P = 0.26). The median WBC counts at initial diagnosis of BCOR/BCORL1mut was significantly lower than that in BCOR/BCORL1wt group (6.79 [IQR, 2.41–29.96] vs. 20.53 [IQR, 3.54–60.80] ×109/L, P = 0.003). Patients with BCOR/BCORL1mut were more frequently categorized within the 2022 ELN adverse-risk group (66.3% vs. 25.7%, P < 0.001), and fewer achieved pre-transplant MRD negativity (61.5% vs. 79.7%, P < 0.001). In the BCOR/BCORL1mut group, the time interval from diagnosis to transplantation was significantly shorter than BCOR/BCORL1wt group as well (6.6 [IQR, 5.6–8.1] months vs. 7.2 [IQR, 5.8–10.0] months, P = 0.04). Additionally, the HCT-CI scores of BCOR/BCORL1mut patients were significantly higher than that of BCOR/BCORL1wt patients (P < 0.001). Other baseline characteristics between the two groups were similar, including donor type, GVHD prophylaxis, and gender matching.
Table 1
Patient characteristics of BCOR/BCORL1mut and BCOR/BCORL1wt group
Characteristics
Total
BCOR/BCORL1mut
BCOR/BCORL1wt
P
(n = 359)
(n = 83)
(n = 276)
Age (years), Mean (Q₁, Q₃)
38 (30, 48)
38 (33, 48)
38 (29, 49)
0.256
Sex, n (%)
   
0.984
Female
177 (49.30)
41 (49.40)
136 (49.28)
 
Male
182 (50.70)
42 (50.60)
140 (50.72)
 
WBC (×109/L), Mean (Q₁, Q₃)
17.33 (2.99, 51.30)
6.79 (2.41, 29.96)
20.53 (3.54, 60.80)
0.003
BM blasts, Mean (Q₁, Q₃)
54.0 (26.0, 77.6)
44.0 (31.0, 72.0)
56.00(24.5, 78.5)
0.629
ELN 2022 risk group, n (%)
   
< 0.001
Favorable
84 (23.40)
9 (10.84)
75 (27.17)
 
Intermediate
149 (41.50)
19 (22.89)
130 (47.10)
 
Adverse
126 (35.10)
55 (66.27)
71 (25.72)
 
r/r AML, n (%)
   
0.152
No
260 (72.42)
55 (66.27)
205 (74.28)
 
Yes
99 (27.58)
28 (33.73)
71 (25.72)
 
Time from diagnosis to transplantation (months), Mean (Q₁, Q₃)
7.1 (5.8, 9.2)
6.6 (5.6, 8.1)
7.2 (5.8, 10.0)
0.037
Pre-transplant status, n (%)
   
< 0.001
CR-MRD (-)
271 (75.49)
51 (61.45)
220 (79.71)
 
CR-MRD (+)
39 (10.86)
18 (21.69)
21 (7.61)
 
NOT-CR
49 (13.65)
14 (16.87)
35 (12.68)
 
HCT-CI score, n (%)
   
< 0.001
≤ 1
317 (88.30)
60 (72.29)
257 (93.12)
 
> 1
42 (11.70)
23 (27.71)
19 (6.88)
 
HSCT type, n (%)
   
0.403
HID
227 (63.23)
49 (59.04)
178 (64.49)
 
MSD
115 (32.03)
28 (33.73)
87 (31.52)
 
URD
17 (4.74)
6 (7.23)
11 (3.99)
 
Prophylaxis of acute GVHD, n (%)
   
0.7
CSA + MTX ± MMF
184 (51.25)
41 (49.40)
143 (51.81)
 
FK506 + MTX ± MMF
175 (48.75)
42 (50.60)
133 (48.19)
 
Gender match, n (%)
   
0.886
Female to Female
61 (16.99)
13 (15.66)
48 (17.39)
 
Female to Male
62 (17.27)
15 (18.07)
47 (17.03)
 
Male to Female
119 (33.15)
30 (36.14)
89 (32.25)
 
Male to Male
117 (32.59)
25 (30.12)
92 (33.33)
 
Abbreviation: WBC, white blood cell; BM, bone marrow; ELN, the European Leukemia Net; r/r AML, relapsed/refractory acute myeloid leukemia; CR, complete remission; MRD, minimal residual disease; HCT-CI, hematopoietic cell transplantation specific comorbidity index; HID, haploidentical donor; MSD, matched sibling donor; URD, unrelated donor; GVHD, graft versus host disease; CSA, cyclosporine A; MTX, methotrexate; MMF, mycophenolate mofetil; FK506, tacrolimus

Survival and relapse

The median follow-up time of the 359 patients was 35.4 (95% CI, 32.3–38.5) months. Three-year OS, DFS, and CIR were 75.5% (95% CI, 70.8-80.4%), 73.3% (95% CI, 68.6-78.3%) and 14.9% (95% CI, 11.2-19.1%), respectively. The median follow-up time for the BCOR/BCORL1mut and BCOR/BCORL1wt groups were 34.8 (95% CI, 25.0-44.6) and 36.0 (95% CI, 32.1–39.9) months, and the maximum follow-up times were 77.0 and 73.3 months, respectively. By the end of follow-up, 17 (20.5%) patients in the BCOR/BCORL1mut group had died, of whom 11 (64.7%) died from relapse, 2 (11.8%) died from grade IV acute GVHD, 3 (17.6%) died from pulmonary infection, and 1(5.9%) died from unknown cause.
The median survival time of the BCOR/BCORL1mut group was 73.4 (95% CI, 33.8–113.0) months, but that of the BCOR/BCORL1wt group was not reached. Furthermore, the three-year OS of the BCOR/BCORL1mut and BCOR/BCORL1wt groups was 76.0% (95% CI, 66.0-87.5%) vs. 75.2% (95% CI, 70.0-80.8%) (HR, 0.92; 95% CI, 0.54–1.57; P = 0.77), respectively (Fig. 2a); the three-year DFS of the BCOR/BCORL1mut and BCOR/BCORL1wt groups was 67.7% (95% CI, 57.0-80.4%) vs. 74.5% (95% CI, 69.4-80.1%) (HR, 1.20; 95% CI, 0.75–1.91; P = 0.46), respectively (Fig. 2b); the three-year CIR of the BCOR/BCORL1mut and BCOR/BCORL1wt groups was 24.0% (95% CI, 14.1-35.4%) vs. 12.6% (95% CI, 8.9-17.0%) (HR, 1.85; 95% CI, 1.04–3.3; P = 0.03), respectively (Fig. 2c); and the three-year NRM of the BCOR/BCORL1mut and BCOR/BCORL1wt groups was 8.3% (95% CI, 3.3-16.2%) vs. 12.9% (95% CI, 9.1-17.2%) (HR, 0.58; 95% CI, 0.25–1.38; P = 0.22), respectively (Fig. 2d).
Fig. 2
Transplant outcomes of acute myeloid leukemia patients with BCOR/BCORL1mut and BCOR/BCORL1wt. (a) Overall survival. (b) Disease-free survival. (c) Cumulative incidence of relapse. (d) Cumulative incidence of non-relapse mortality
Bild vergrößern
Clinical outcomes of the four groups, including BCORwt/BCORL1wt, BCORmut/BCORL1wt, BCORwt/BCORL1mut, and BCORmut/BCORL1mut patients, were also analyzed. No significant differences were observed in OS, DFS, CIR, or NRM among any of these groups. However, the CIR was significantly higher in BCORmut/BCORL1wt patients compared to BCORwt/BCORL1wt patients (3-year CIR: 29.0% [95% CI, 15.0-44.7%] vs. 12.6% [95% CI, 8.9-17.0%], HR, 2.44 [95% CI, 1.28–4.65], P = 0.007), but no significant difference was found between BCORmut/BCORL1wt and BCORwt/BCORL1mut group (3-year CIR: 29.0% [95% CI, 15.0-44.7%] vs. 20.0% [95% CI, 5.8%-4.3%], HR, 0.53 [95% CI, 0.18–1.59], P = 0.26) (Fig S1).
We also compared the prognostic differences between BCOR/BCORL1mut and BCOR/BCORL1wt groups across different MRD statuses, 2022 ELN risk stratifications, and donor types. Notably, we observed that BCOR/BCORL1mut in patients undergoing matched sibling donor (MSD) transplantation were associated with an increased relapse rate (HR, 3.48; 95% CI, 1.41–8.60; P = 0.01) but lower NRM (HR, 0; 95% CI, 0-Inf; P = 0.02) after allo-HSCT. No difference between other conditions was found (Table S2).

Risk factors analysis

As shown by the results above, we further conducted univariate and multivariate analyses in 359 patients to assess whether BCOR/BCORL1mut was an independent risk factor for CIR after transplantation (Table 2). These results showed that BCOR/BCORL1mut was associated with increased CIR after allo-HSCT in univariate analysis (HR, 1.85; 95% CI, 1.04–3.3; P = 0.03) but was not an independent risk factor in multivariate analysis (HR, 1.31; 95% CI, 0.59–2.87; P = 0.51). Additionally, pre-transplant MRD positivity (HR, 2.56; 95%CI, 1.01–6.46; P = 0.04) and not-CR (HR, 5.63; 95% CI, 2.69–11.82; P < 0.001) independently predicted an unfavorable outcome for CIR. Univariate and multivariate analysis for OS and DFS were also conducted and detailed results are listed in Table 2.
Table 2
Univariate and multivariate analysis in 359 AML patients
OS
Univariate analysis
Multivariate analysis
HR (95% CI) p-value
HR (95% CI) p-value
Age (years)
  
≥ 50 vs. < 50
1.64 (1.03–2.60) 0.04
1.65 (1.03–2.66) 0.04
Sex
  
Female vs. male
1.25 (0.82–1.93) 0.30
 
WBC (×109/L)
  
≥ 100 vs. < 100
0.97 (0.50–1.87) 0.92
 
Bone marrow blasts
  
≥ 50% vs. < 50%
0.78 (0.51–1.20) 0.26
 
BCOR/BCORL1 mutation
  
Yes vs. no
0.92 (0.54–1.57) 0.77
 
2022 ELN
  
Intermediate vs. favorable
2.36 (1.21–4.62) 0.01
2.32 (1.18–4.56) 0.01
Adverse vs. favorable
2.61 (1.32–5.16) 0.01
1.82 (0.89–3.70) 0.10
Pre-HSCT status
  
CR-MRD (+) vs. CR-MRD (-)
1.90 (1.03–3.54) 0.04
1.68 (0.88–3.20) 0.11
NOT-CR vs. CR-MRD (-)
3.28 (2.00–5.35) < 0.001
3.06 (1.82–5.15) < 0.001
HCT-CI score
  
> 1 vs. ≤ 1
1.56 (0.86–2.82) 0.14
 
Donor type
  
HID vs. MSD
0.96 (0.61–1.51) 0.86
 
URD vs. MSD
0.45 (0.11–1.87) 0.27
 
Acute GVHD prophylaxis
  
FK506 + MTX ± MMF vs. CSA + MTX ± MMF
1.05 (0.69–1.61) 0.81
 
DFS
  
Age (years)
  
≥ 50 vs. < 50
1.58 (1.01–2.46) 0.04
1.52 (0.97–2.39) 0.07
Sex
  
Female vs. male
1.36 (0.90–2.04) 0.14
 
WBC (×109/L)
  
≥ 100 vs. < 100
0.83 (0.43–1.61) 0.59
 
Bone marrow blasts
  
≥ 50% vs. < 50%
0.74 (0.49–1.11) 0.14
 
BCOR/BCORL1 mutation
  
Yes vs. no
1.19 (0.75–1.91) 0.46
 
2022 ELN
  
Intermediate vs. favorable
1.97 (1.05–3.70) 0.03
1.94 (1.03–3.65) 0.04
Adverse vs. favorable
2.78 (1.49–5.19) 0.001
1.99 (1.04–3.82) 0.04
Pre-HSCT status
  
CR-MRD (+) vs. CR-MRD (-)
1.96 (1.09–3.53) 0.03
1.66 (0.90–3.06) 0.10
NOT-CR vs. CR-MRD (-)
3.28 (2.05–5.24) < 0.001
2.88 (1.75–4.72) < 0.001
HCT-CI score
  
> 1 vs. ≤ 1
1.33 (0.74–2.38) 0.34
 
Donor type
  
HID vs. MSD
0.85 (0.56–1.30) 0.45
 
URD vs. MSD
0.36 (0.09–1.52) 0.17
 
Acute GVHD prophylaxis
  
FK506 + MTX ± MMF vs. CSA + MTX ± MMF
1.10 (0.73–1.65) 0.65
 
CIR
  
Age (years)
  
≥ 50 vs. < 50
1.00 (0.56–2.01) 0.87
 
Sex
  
Female vs. male
1.41 (0.82–2.43) 0.22
 
WBC (×10*9/L)
  
≥ 100 vs. < 100
0.74 (0.30–1.86) 0.52
 
Bone marrow blasts
  
≥ 50% vs. < 50%
0.48 (0.27–0.85) 0.01
0.86 (0.35–2.08) 0.73
BCOR/BCORL1 mutation
  
Yes vs. no
1.85 (1.04–3.3) 0.03
1.31 (0.59–2.87) 0.51
2022 ELN
  
Intermediate vs. favorable
1.42 (0.59–3.39) 0.43
1.44 (0.54–3.82) 0.46
Adverse vs. favorable
3.39 (1.51–7.59) 0.003
1.27 (0.49–3.29) 0.63
Pre-HSCT status
  
CR-MRD (+) vs. CR-MRD (-)
2.30 (1.04–5.08) < 0.001
2.56 (1.01–6.46) 0.04
NOT-CR vs. CR-MRD (-)
4.48 (2.42–8.28) < 0.001
5.63 (2.69–11.82) < 0.001
HCT-CI score
  
> 1 vs. ≤ 1
1.02 (0.43–2.43) 0.96
 
Donor type
  
HID vs. MSD
0.90 (0.52–1.58) 0.72
 
URD vs. MSD
0.35 (0.05–2.61) 0.30
 
Acute GVHD prophylaxis
  
FK506 + MTX ± MMF vs. CSA + MTX ± MMF
0.87 (0.51–1.49) 0.61
 
Abbreviation: OS, overall survival; DFS, disease-free survival; CIR, cumulative incidence of relapse; WBC, white blood cell; r/r AML, relapsed/refractory acute myeloid leukemia; ELN, the European Leukemia Net; HSCT, hematopoietic stem cell transplantation; CR, complete remission; MRD, minimal residual disease; HCT-CI, hematopoietic cell transplantation specific comorbidity index; HID, haploidentical donor; MSD, matched sibling donor; URD, unrelated donor; FK506, tacrolimus; CSA, cyclosporine A; MTX, methotrexate; MMF, mycophenolate mofetil

Impact of mutation characteristics on transplant outcomes

We investigated whether the type and location of BCOR/BCORL1mut influenced transplant outcomes. Strikingly, there was no significant difference in DFS or CIR between patients with frameshift or other types of mutation (Fig. 3a, b), and comparable results were also found for the location of mutations (Fig. 3c, d). The three-year OS, DFS, and CIR of patients with frameshift mutations and other types of mutations were 88.1% (95% CI, 76.3-100.0%) vs. 71.1% (95% CI, 58.6-86.2%) (HR, 0.58; 95% CI, 0.19–1.78; P = 0.34), 74.1% (95% CI, 57.2-96.0%) vs. 64.5% (95% CI, 51.6-80.8%) (HR, 0.75; 95% CI, 0.29–1.89; P = 0.53) and 22.2% (95% CI, 6.9-42.8%) vs. 24.9% (95% CI, 12.9-38.9%) (HR, 0.91; 95% CI, 0.32–2.60; P = 0.86), respectively. The three-year OS, DFS, and CIR of patients with C-terminal domains mutations and other locations of mutations were 100.0% (95% CI, 100.0%-100.0%) vs. 74.9% (95% CI, 64.6-86.8%) (HR, 0; 95% CI, 0-InF; P = 0.33), 100.0% (95% CI, 100.0%-100.0%) vs. 66.2% (95% CI, 55.2-79.3%) (HR, 0; 95% CI, 0-InF, P = 0.25), and 0 vs. 25.1% (95% CI, 14.8-36.7%) (HR, 0; 95% CI, 0-InF; P = 0.33), respectively (Table 3).
Fig. 3
Impact of BCOR and BCORL1 mutation characteristics on transplant outcomes. (a) Disease-free survival (DFS) and (b) cumulative incidence of relapse (CIR) in patients with frameshift and other type of mutation. DFS (c) and CIR (d) in patients with alteration in C-terminal domain and other locations
Bild vergrößern
Table 3
Three-year OS, DFS and CIR of patients with different mutation type and location in BCOR/BCORL1mut group
 
Mutation type
Mutation location
Frame shift
other types
C- terminal domains
other locations
OS
 
Three-year OS
(95%CI)
88.1%
(76.3-100.0%)
71.1%
(58.6-86.2%)
100.0%
(100.0%-100.0%)
74.9%
(64.6-86.8%)
HR (95%CI)
p-value
0.58 (0.19–1.78)
P = 0.34
0 (0-InF)
P = 0.33
DFS
 
Three-year DFS
(95%CI)
74.1%
(57.2-96.0%)
64.5%
(51.6-80.8%)
100.0%
(100.0%-100.0%)
66.2%
(55.2-79.3%)
HR (95%CI)
p-value
0.75 (0.29–1.89)
P = 0.53
0 (0-InF)
P = 0.25
CIR
 
Three-year CIR
(95%CI)
22.2%
(6.9-42.8%)
24.9%
(12.9-38.9%)
0
25.1%
(14.8-36.7%)
HR (95%CI)
p-value
0.91 (0.32–2.60)
P = 0.86
0 (0-InF)
P = 0.33
Abbreviation: OS, overall survival; DFS, disease-free survival; CIR, cumulative incidence of relapse

Discussion

BCOR and BCORL1 are key transcription factors and function as components of PRC1.1, a noncanonical PRC1 complex, that monoubiquitinates histone 2 A at lysine 119 and mediates transcriptional repression [23]. BCOR mutations have been widely reported in patients with hematologic malignancies and mesenchymal tumors [3]. For AML, BCOR/BCORL1mut ranges from less than 10% and occurs primarily in cytogenetically normal AML (CN-AML) [4]. In the 2022 ELN risk stratification, BCOR mutations are classified as an adverse prognostic marker, suggesting that AML patients with BCOR mutations are more likely to be classified into the adverse-risk group. For these patients, the importance of allo-HSCT is heightened [11]. BCOR/BCORL1mut has also been reported with a reduced DFS, but this did not significantly affect OS in an unselected cohort [4, 7, 8]. However, the role of BCOR/BCORL1mut in allo-HSCT remains unclear. In this study, we described the clinical characteristics and transplant outcomes of eighty-three BCOR/BCORL1mut AML patients and compared them to CN-AML. To our knowledge, this is the largest study to examine the influence of BCOR/BCORL1mut in a transplant cohort.
The prevalence of BCOR and BCORL1 mutations in our patients were 6.4% and 4.3%, respectively, consistent with previous studies [6, 7, 14]. We observed lower WBC counts at diagnosis in BCOR- and BCORL1-mutated patients, and previous studies have also suggested that AML patients with BCOR mutation are characterized by excessive proliferation of blasts and reduced leukocyte hyperplasia [12, 13, 24]. We also observed that BCOR-mutated AML showed a lower CR rate pre-transplantation. Japan researchers have also reported a significantly lower CR rate (47.4% vs. 71.8%, P = 0.0357) in cases with BCOR/BCORL1mut than in cases with BCOR/BCORL1wt [5]. In mechanistic terms, research has demonstrated that BCOR knockout cell lines exhibit reductions in sensitivity to chemotherapy with Idarubicin and Ara-C in mice model [25]. A lower CR rate also contributes to a shorter interval to transplantation in BCOR/BCORL1mut patients.
In the present study, we found that the predominant type of BCOR mutation was frameshift mutations, followed by missense mutation; and missense mutation accounted for the majority of BCORL1 mutations. These results coincide with those reported previously [4, 8, 26]. The therapeutic impact of mutation characteristics has not been totally established. Abuhadra et al. reported a median OS of 10.9 months for patients with frameshift mutations, compared to 50.4 months for patients with other types of mutations (P = 0.03) [26], though we did not find a significant difference in transplant outcomes between cases with frameshift mutations and those with other mutation types. Mutation location has also been reported to be associated with clinical outcomes [26]. At present, four functional domains of the BCOR gene were well-established, including BBD, AF9BD, ANK repeats and PUFD [1], and the BCORL1 gene is characterized by the CBS, 2 LXXLL, ANK repeats, and PUFD [1]. BCOR altered at C-terminal tandem ANK was reported to have a lower OS than mutations at other positions [26]. In this study, the distribution of mutation sites was quite diverse and most mutations were located out of functional domains. Our analysis showed that mutations occurring at the C-terminal domain had no significant effect on transplant outcomes. These results indicate that allo-HSCT may overcome the prognostic impact of mutation type and location.
AML can stem from multiple mutations [10, 11, 27, 28]. BCOR mutations have been reported to be mutually exclusive with NPM1 mutations, and associated with a lower incidence of FLT3-ITD mutations, and to have an association with DNMT3A and RUNX1 mutations [4, 5, 7], which are all consistent with our findings except for the higher frequency of the co-mutation rate of FLT3 gene in our cohort. The fact that our subjects were exclusively Chinese may explain this, however, because of the combined effects of genetic background, disease distribution, and environmental factors. DNMT3A mutation is associated with poor survival outcomes, and co-mutation of BCOR and DNMT3A has been widely reported [2931]. Researchers speculated that these two mutations may act synergistically to induce AML by interfering with epigenetic mechanisms, though more research is needed to confirm [32].
Most BCOR-mutated AML patients carry normal karyotypes [4]. Several studies have compared BCOR-mutated AML with CN-AML in chemo-based cohorts and found a shorter OS in BCOR-mutated AML patients. In addition, they have also indicated that allo-HSCT can reverse the adverse outcomes caused by BCOR mutations [33]. However, the impact of BCOR/BCORL1mut on transplant outcomes has yet to be reported specifically. In our present study, the OS, DFS, and NRM of BCOR/BCORL1mut patients were quite similar to outcomes of BCOR/BCORL1wt patients. Notably, BCOR/BCORL1mut patients had a higher CIR after transplantation compared to BCOR/BCORL1wt patients, suggesting a potential impact of BCOR/BCORL1 mutations on relapse. We also observed a slightly higher NRM in the BCOR/BCORL1wt group, which might explain the equivalent DFS between BCOR/BCORL1mut and BCOR/BCORL1wt patients, despite the increased CIR in the BCOR/BCORL1mut group. Although the difference in NRM between the two groups was not statistically significant, the interval from diagnosis to transplantation, the number of chemotherapy cycles, and immune status may all be potential factors that influence NRM. Although further analysis showed that BCOR/BCORL1mut was not an independent risk factor for CIR, it was strongly related to a higher CIR post-transplantation and thus a potentially beneficial future research direction would be to perform a large registry study to observe transplant patients with BCOR/BCORL1mut prospectively.
There are limitations to this study that should be acknowledged. First, this was a single-center and retrospective study. Second, we could not compare the efficacy of chemotherapy and transplantation due to the lack of chemo-based cohort.
In summary, we are the first to our knowledge to report the comparable transplant outcomes of BCOR- and BCORL1-mutated AML with CK-AML. BCOR- and BCORL1-mutated AML tend to have a higher tendency to relapse, however, which requires further investigation in large-scale multicenter studies.

Acknowledgements

The authors thank AiMi Academic Services (www.​aimieditor.​com) for English language editing and review services.

Declarations

All cases were included in the National Longitudinal Cohort of Hematological Diseases (NICHE, NCT04645199) which was approved by the Ethics Committee of IHCAMS. All patients provided informed consent for clinical information for scientific research at the time of first admission.
All the authors consent for publication.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc-nd/​4.​0/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

download
DOWNLOAD
print
DRUCKEN
Titel
Impact of BCOR/BCORL1 mutation on outcomes of allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia patients
Verfasst von
YunXia Zhou
Haixiao Zhang
Xinhui Zheng
Rongli Zhang
Xin Chen
Qiaoling Ma
Donglin Yang
Jialin Wei
Aiming Pang
Yi He
Sizhou Feng
Mingzhe Han
Weihua Zhai
Erlie Jiang
Publikationsdatum
09.04.2025
Verlag
Springer Berlin Heidelberg
Erschienen in
Annals of Hematology / Ausgabe 5/2025
Print ISSN: 0939-5555
Elektronische ISSN: 1432-0584
DOI
https://doi.org/10.1007/s00277-025-06346-6

Electronic supplementary material

Below is the link to the electronic supplementary material.
1.
Zurück zum Zitat Sportoletti P, Sorcini D, Falini B (2021) BCOR gene alterations in hematologic diseases. Blood 138:2455–2468CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Huynh KD, Fischle W, Verdin E, Bardwell VJ (2000) BCoR, a novel corepressor involved in BCL-6 repression. Genes Dev 14:1810–1823CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Astolfi A, Fiore M, Melchionda F, Indio V, Bertuccio SN, Pession A (2019) BCOR involvement in cancer. Epigenomics 11:835–855CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Grossmann V, Tiacci E, Holmes AB, Kohlmann A, Martelli MP, Kern W, Spanhol-Rosseto A, Klein HU, Dugas M, Schindela S, Trifonov V, Schnittger S, Haferlach C, Bassan R, Wells VA, Spinelli O, Chan J, Rossi R, Baldoni S, De Carolis L, Goetze K, Serve H, Peceny R, Kreuzer KA, Oruzio D, Specchia G, Di Raimondo F, Fabbiano F, Sborgia M, Liso A, Farinelli L, Rambaldi A, Pasqualucci L, Rabadan R, Haferlach T, Falini B (2011) Whole-exome sequencing identifies somatic mutations of BCOR in acute myeloid leukemia with normal karyotype. Blood 118:6153–6163CrossRefPubMed
5.
Zurück zum Zitat Terada K, Yamaguchi H, Ueki T, Usuki K, Kobayashi Y, Tajika K, Gomi S, Kurosawa S, Saito R, Furuta Y, Miyadera K, Tokura T, Marumo A, Omori I, Sakaguchi M, Fujiwara Y, Yui S, Ryotokuji T, Arai K, Kitano T, Wakita S, Fukuda T, Inokuchi K (2018) Usefulness of BCOR gene mutation as a prognostic factor in acute myeloid leukemia with intermediate cytogenetic prognosis. Genes Chromosomes Cancer 57:401–408CrossRefPubMed
6.
Zurück zum Zitat Nazha A, Zarzour A, Al-Issa K, Radivoyevitch T, Carraway HE, Hirsch CM, Przychodzen B, Patel BJ, Clemente M, Sanikommu SR, Kalaycio M, Maciejewski JP, Sekeres MA (2016) The complexity of interpreting genomic data in patients with acute myeloid leukemia. Blood Cancer J 6:e510CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Damm F, Chesnais V, Nagata Y, Yoshida K, Scourzic L, Okuno Y, Itzykson R, Sanada M, Shiraishi Y, Gelsi-Boyer V, Renneville A, Miyano S, Mori H, Shih LY, Park S, Dreyfus F, Guerci-Bresler A, Solary E, Rose C, Cheze S, Prébet T, Vey N, Legentil M, Duffourd Y, de Botton S, Preudhomme C, Birnbaum D, Bernard OA, Ogawa S, Fontenay M, Kosmider O (2013) BCOR and BCORL1 mutations in myelodysplastic syndromes and related disorders. Blood 122:3169–3177CrossRefPubMed
8.
Zurück zum Zitat Baranwal A, Gurney M, Basmaci R, Katamesh B, He R, Viswanatha DS, Greipp P, Foran J, Badar T, Murthy H, Yi CA, Palmer J, Mangaonkar AA, Patnaik MM, Litzow MR, Hogan WJ, Begna K, Gangat N, Tefferi A, Al-Kali A, Shah MV, Alkhateeb HB (2024) Genetic landscape and clinical outcomes of patients with BCOR mutated myeloid neoplasms. Haematologica 109:1779–1791PubMedPubMedCentral
9.
Zurück zum Zitat Song GY, Kim T, Ahn SY, Jung SH, Kim M, Yang DH, Lee JJ, Choi SH, Kim MY, Jung CW, Jang JH, Kim HJ, Moon JH, Sohn SK, Won JH, Park SK, Kim SH, Zhang Z, Ahn JS, Kim HJ, Kim DDH (2022) Allogeneic hematopoietic cell transplantation can overcome the adverse prognosis indicated by secondary-type mutations in de Novo acute myeloid leukemia. Bone Marrow Transpl 57:1810–1819CrossRef
10.
Zurück zum Zitat Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S, O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell PJ (2016) Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med 374:2209–2221CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Döhner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, Ebert BL, Fenaux P, Godley LA, Hasserjian RP, Larson RA, Levine RL, Miyazaki Y, Niederwieser D, Ossenkoppele G, Röllig C, Sierra J, Stein EM, Tallman MS, Tien HF, Wang J, Wierzbowska A, Löwenberg B (2022) Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood 140:1345–1377CrossRefPubMed
12.
Zurück zum Zitat Cao Q, Gearhart MD, Gery S, Shojaee S, Yang H, Sun H, Lin DC, Bai JW, Mead M, Zhao Z, Chen Q, Chien WW, Alkan S, Alpermann T, Haferlach T, Müschen M, Bardwell VJ, Koeffler HP (2016) BCOR regulates myeloid cell proliferation and differentiation. Leukemia 30:1155–1165CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Kelly MJ, So J, Rogers AJ, Gregory G, Li J, Zethoven M, Gearhart MD, Bardwell VJ, Johnstone RW, Vervoort SJ, Kats LM (2019) Bcor loss perturbs myeloid differentiation and promotes leukaemogenesis. Nat Commun 10:1347CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Li M, Collins R, Jiao Y, Ouillette P, Bixby D, Erba H, Vogelstein B, Kinzler KW, Papadopoulos N, Malek SN (2011) Somatic mutations in the transcriptional corepressor gene BCORL1 in adult acute myelogenous leukemia. Blood 118:5914–5917CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Niederwieser D, Baldomero H, Bazuaye N, Bupp C, Chaudhri N, Corbacioglu S, Elhaddad A, Frutos C, Galeano S, Hamad N, Hamidieh AA, Hashmi S, Ho A, Horowitz MM, Iida M, Jaimovich G, Karduss A, Kodera Y, Kröger N, de Péffault R, Lee JW, Martínez-Rolón J, Pasquini MC, Passweg J, Paulson K, Seber A, Snowden JA, Srivastava A, Szer J, Weisdorf D, Worel N, Koh MBC, Aljurf M, Greinix H, Atsuta Y, Saber W (2022) One and a half million hematopoietic stem cell transplants: continuous and differential improvement in worldwide access with the use of non-identical family donors. Haematologica 107:1045–1053CrossRefPubMed
16.
Zurück zum Zitat D’Souza A, Fretham C, Lee SJ, Arora M, Brunner J, Chhabra S, Devine S, Eapen M, Hamadani M, Hari P, Pasquini MC, Perez W, Phelan RA, Riches ML, Rizzo JD, Saber W, Shaw BE, Spellman SR, Steinert P, Weisdorf DJ, Horowitz MM (2020) Current use of and trends in hematopoietic cell transplantation in the united States. Biol Blood Marrow Transpl 26:e177–e182CrossRef
17.
Zurück zum Zitat Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, Advani R, Ghielmini M, Salles GA, Zelenetz AD, Jaffe ES (2016) The 2016 revision of the world health organization classification of lymphoid neoplasms. Blood 127:2375–2390CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Kumar RD, Searleman AC, Swamidass SJ, Griffith OL, Bose R (2015) Statistically identifying tumor suppressors and oncogenes from pan-cancer genome-sequencing data. Bioinformatics 31:3561–3568CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Schoemans HM, Lee SJ, Ferrara JL, Wolff D, Levine JE, Schultz KR, Shaw BE, Flowers ME, Ruutu T, Greinix H, Holler E, Basak G, Duarte RF, Pavletic SZ (2018) EBMT-NIH-CIBMTR task force position statement on standardized terminology & guidance for graft-versus-host disease assessment. Bone Marrow Transpl 53:1401–1415CrossRef
20.
Zurück zum Zitat Jagasia MH, Greinix HT, Arora M, Williams KM, Wolff D, Cowen EW, Palmer J, Weisdorf D, Treister NS, Cheng GS, Kerr H, Stratton P, Duarte RF, McDonald GB, Inamoto Y, Vigorito A, Arai S, Datiles MB, Jacobsohn D, Heller T, Kitko CL, Mitchell SA, Martin PJ, Shulman H, Wu RS, Cutler CS, Vogelsang GB, Lee SJ, Pavletic SZ, Flowers ME (2015) National institutes of health consensus development project on criteria for clinical trials in chronic Graft-versus-Host disease: I. The 2014 diagnosis and staging working group report. Biol Blood Marrow Transpl 21:389–401e381CrossRef
21.
Zurück zum Zitat Srinivasan RS, de Erkenez AC, Hemenway CS (2003) The mixed lineage leukemia fusion partner AF9 binds specific isoforms of the BCL-6 corepressor. Oncogene 22:3395–3406CrossRefPubMed
22.
Zurück zum Zitat Uckelmann HJ, Armstrong SA (2020) Chromatin complexes maintain Self-Renewal of myeloid progenitors in AML: opportunities for therapeutic intervention. Stem Cell Rep 15:6–12CrossRef
23.
Zurück zum Zitat Di Carlo V, Mocavini I, Di Croce L (2019) Polycomb complexes in normal and malignant hematopoiesis. J Cell Biol 218:55–69CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Tara S, Isshiki Y, Nakajima-Takagi Y, Oshima M, Aoyama K, Tanaka T, Shinoda D, Koide S, Saraya A, Miyagi S, Manabe I, Matsui H, Koseki H, Bardwell VJ, Iwama A (2018) Bcor insufficiency promotes initiation and progression of myelodysplastic syndrome. Blood 132:2470–2483CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Honda A, Koya J, Yoshimi A, Miyauchi M, Taoka K, Kataoka K, Arai S, Kurokawa M (2021) Loss-of-function mutations in BCOR contribute to chemotherapy resistance in acute myeloid leukemia. Exp Hematol 101–102:42–48e11CrossRefPubMed
26.
Zurück zum Zitat Abuhadra N, Mukherjee S, Al-Issa K, Adema V, Hirsch CM, Advani A, Przychodzen B, Makhoul A, Awada H, Maciejewski JP, Sekeres MA, Nazha A (2019) BCOR and BCORL1 mutations in myelodysplastic syndromes (MDS): clonal architecture and impact on outcomes. Leuk Lymphoma 60:1587–1590CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat De Kouchkovsky I, Abdul-Hay M (2016) Acute myeloid leukemia: a comprehensive review and 2016 update. Blood Cancer J 6:e441CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Kayser S, Levis MJ (2023) The clinical impact of the molecular landscape of acute myeloid leukemia. Haematologica 108:308–320CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Ley TJ, Ding L, Walter MJ, McLellan MD, Lamprecht T, Larson DE, Kandoth C, Payton JE, Baty J, Welch J, Harris CC, Lichti CF, Townsend RR, Fulton RS, Dooling DJ, Koboldt DC, Schmidt H, Zhang Q, Osborne JR, Lin L, O’Laughlin M, McMichael JF, Delehaunty KD, McGrath SD, Fulton LA, Magrini VJ, Vickery TL, Hundal J, Cook LL, Conyers JJ, Swift GW, Reed JP, Alldredge PA, Wylie T, Walker J, Kalicki J, Watson MA, Heath S, Shannon WD, Varghese N, Nagarajan R, Westervelt P, Tomasson MH, Link DC, Graubert TA, DiPersio JF, Mardis ER, Wilson RK (2010) DNMT3A mutations in acute myeloid leukemia. N Engl J Med 363:2424–2433CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Brunetti L, Gundry MC, Goodell MA (2017) DNMT3A in leukemia. Cold Spring Harb Perspect Med 7
31.
Zurück zum Zitat Khrabrova DA, Yakubovskaya MG, Gromova ES (2021) AML-Associated mutations in DNA methyltransferase DNMT3A. Biochem (Mosc) 86:307–318CrossRef
32.
Zurück zum Zitat Tiacci E, Grossmann V, Martelli MP, Kohlmann A, Haferlach T, Falini B (2012) The corepressors BCOR and BCORL1: two novel players in acute myeloid leukemia. Haematologica 97:3–5CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Zhang A, Liu Y, Wei S, Gong B, Zhou C, Wang Y, Wei H, Mi Y, Wang J (2020) BCOR mutations in acute myeloid leukemia: clonal evolution and prognosis. Blood 136:4–4CrossRef

Kompaktes Leitlinien-Wissen Innere Medizin (Link öffnet in neuem Fenster)

Mit medbee Pocketcards schnell und sicher entscheiden.
Leitlinien-Wissen kostenlos und immer griffbereit auf ihrem Desktop, Handy oder Tablet.

Neu im Fachgebiet Innere Medizin

Podcast

Depression: Die Zeit bis zum Therapieplatz sinnvoll nutzen

Die Zahl der diagnostizierten Depressionen steigt – gleichzeitig fehlen flächendeckend Therapieplätze. Für Betroffene ist die Hausarztpraxis eine erste Anlaufstelle. Doch wie kann dort der passende Raum entstehen? Allgemeinmedizinerin Prof. Dr. med. Anne Simmenroth und Psychologin Maike Krauthausen berichten, wie niederschwellig Hilfe angeboten werden kann.

Zeitschrift für Allgemeinmedizin, DEGAM

FLT3-Hemmung wirkt auch bei FLT3-ITD-negativer AML

Ergebnisse der Phase-II-Studie QUIWI nach längerem Follow-up sprechen weiter dafür, dass auch Personen mit neu diagnostizierter FLT3-ITD-negativer AML von einer Erweiterung der Standardchemotherapie um den zielgerichteten Wirkstoff Quizartinib profitieren.

Knoten unter der Haut bei Diabetes-Patientin – Kennen Sie die Diagnose?

Wiederholte Insulininjektionen an derselben Stelle können in eine Lipohypertrophie münden. Eine andere, ähnlich aussehende Folgekomplikation, die nicht von selbst verschwindet, ist seltener und weniger bekannt. Eine Kasuistik.

Deutliche Rezidivangst bei jeder siebten Brustkrebspatientin

Fünf Jahre nach Diagnose eines frühen Brusttumors leidet jede siebte Langzeitüberlebende unter ausgeprägter Rezidivangst. Besonders betroffen sind Frauen mit Nebenwirkungen der endokrinen Therapie oder ohne eine solche Behandlung.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

Bildnachweise
Die Leitlinien für Ärztinnen und Ärzte, ZFA TALKS - Depression/© (M) seb_ra / Getty Images / iStock (Symbolbild mit Fotomodell) Logo: Springer Medizin Verlag GmbH, Histopathologischer Befund bei akuter myeloischer Leukämie/© National Cancer Institute/cancer.gov, Frau spritzt sich Insulin per Pen/© Burcu Saritas / Stock.adobe.com (Symbolbild mit Fotomodell), Ältere Frau fasst sich an die Brust/© Jelena Stanojkovic / Stock.adobe.com (Symbolbild mit Fotomodell)