Original Article

Split Viewer

Blood Res 2022; 57(3):

Published online September 30, 2022

https://doi.org/10.5045/br.2022.2022104

© The Korean Society of Hematology

Relevance of flow cytometric categorization and end-of-induction measurable residual disease assessment in pediatric and adult T-lymphoblastic leukemia patients

Karthik Bommannan1, Jhansi Rani Arumugam1, Venkatraman Radhakrishnan2, Jayachandran Perumal Kalaiyarasi2, Parathan Karunakaran2, Nikita Mehra2, Tenali Gnana Sagar2, Shirley Sundersingh1

Departments of 1Oncopathology and 2Medical Oncology, Cancer Institute (W.I.A.), Adyar, India

Correspondence to : Karthik Bommannan, M.D., D.M.
Department of Oncopathology, Cancer Institute (W.I.A), Adyar, Chennai 600020, India
E-mail: bkkb87@gmail.com

*This study was supported by a grant from Indian Council of Medical Research (ICMR) and the Indian Childhood Collaborative Leukemia (ICiCLe) project of the National Cancer Grid (NCG).

Received: May 16, 2022; Revised: June 15, 2022; Accepted: June 21, 2022

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Background
T-lymphoblastic leukemia (T-ALL) patients expressing myeloid/stem cell antigens are classified as early T-cell precursor lymphoblastic leukemia (ETP-ALL) or near-ETP-ALL.
Methods
Clinico-laboratory profiles, flow cytometric end-of-induction measurable residual disease (EOI-MRD), and survival of treatment naïve T-ALL patients were analyzed according to their immunophenotypic subtypes.
Results
Among 81 consecutive T-ALL patients diagnosed, 21% (N=17) were ETP-ALL and 19% (N=15) were near-ETP-ALL. EOI-MRD was detectable in 39% of the 59 samples tested (31.6% of pediatric samples and 52.4% of adult samples). The frequency of EOI-MRD positivity was significantly higher among ETP-ALL (75%, P=0.001) and near-ETP-ALL (71%, P=0.009) patients compared to that in conventional-T-ALL (con-T-ALL) patients (22.5%). CD8 (P=0.046) and CD38 (P=0.046) expressions were significantly upregulated in the EOI blasts of con-T-ALL and ETP-ALL samples, respectively. The 2-year rates of overall (OS), relapse-free (RFS), and event-free survival (EFS) among the T-ALL patients (pediatric vs. adult) was 79.5% vs. 39.8% (P<0.001), 84.3% vs. 60.4% (P=0.026), and 80.3% vs. 38% (P<0.001), respectively. Univariate analysis revealed that 2-year EFS and RFS of pediatric T-ALL patients was independent of T-ALL subtype and was influenced only by EOI-MRD status. However, 2-year OS, RFS, and EFS among adult T-ALL patients were EOI-MRD independent and influenced only by the near-ETP-ALL phenotype.
Conclusion
Two-year survival among pediatric and adult T-ALL patients is attributed to EOI-MRD status and near-ETP-ALL phenotype, respectively.

Keywords Measurable residual disease, Flow cytometry, T-lineage acute lymphoblastic leuke mia, ETP-ALL, Near-ETP-ALL

T-lymphoblastic leukemia (T-ALL) comprises 15% of pediatric and 25% of adult acute lymphoblastic leukemia patients [1]. First described by Coustan-Smith et al. [2] in 2009, ‘early T-cell precursor lymphoblastic leukemia’ (ETP-ALL) is a subtype of T-ALL in which T-lymphoblasts express myeloid/stem cell-associated antigens in the absence of CD1a, CD5, and CD8 expression. ‘Near-ETP-ALL’ is a T-ALL subtype recognized by the World Health Organization (WHO) in 2017. In this subtype, T-lymphoblasts meet all immunophenotype criteria for ETP-ALL, except for having significant CD5 expression [1-3]. Clinical and laboratory characteristics of pediatric and adult ETP-ALL patients have been documented. However, data of near-ETP-ALL patients are limited [4, 5]. Also, the role of other lineage-specific and non-lineage-specific antigens that could distinguish between T-ALL subtypes is unknown.

Flow cytometric measurable residual disease (FCM-MRD) assessment is important for the risk-adapted management of B-lymphoblastic leukemia (B-ALL) patients. However, FCM-MRD-based treatment decisions are not yet part of the management protocols for T-ALL patients. This reflects the limited availability of literature on FCM-MRD in T-ALL. Most of the available publications have included both ETP-ALL and near-ETP-ALL as a common category for data analysis [4, 6-9].

To the best of our knowledge, data comparing age group specific clinico-laboratory profiles across the immunophenotypic subcategories of T-ALL patients are still lacking. Presently, we share our experience regarding clinico-laboratory profiles, end-of-induction (EOI) FCM-MRD, and 2-year survival outcomes of pediatric and adult T-ALL patients immunophenotypically subclassified according to the WHO 2017 guidelines.

This retrospective study was approved by our Institute’s ethics committee. All treatment naïve T-ALL patients diagnosed between December 2017 to March 2020 were included. T-ALL was diagnosed by morphologic evaluation of peripheral blood (PB) and bone marrow (BM) aspiration smears, followed by a 10-color FCM analysis (Supplementary Table 1). Hyperleukocytosis was defined as ≥100×109/L leukocytes in PB [7]. Pediatric (age ≤18 yr) and adult patients were treated with the Indian Collaborative Childhood Leukemia group (high risk-arm) and Berlin-Frankfurt-Muenster (BFM) 95 protocols, respectively [10]. Treatment protocols were not influenced by T-ALL immunophenotype subcategory or end-of-induction measurable residual disease (EOI-MRD) status. During induction, an absolute PB blast count ≥1,000 cells/µL on day 8 of treatment was considered ‘day 8 blasts not cleared’ (D8BNC) status [11, 12].

Diagnostic flow cytometry

BM samples were processed using our previously described ‘lyse-stain-wash’ protocol [13]. A minimum of 100,000 events were acquired per tube using a Beckman Coulter Navios EX flow cytometer. Generated list-mode data (LMD) files were analyzed with Kaluza (Version 2.0) software (Beckman Coulter) using our in-house developed analysis templates. The antigen expression profile was reported according to the Associazione Italiana Ematologia Oncologia Pediatrica-BFM (AIEOP-BFM) 2016 recommendations [14]. The expression intensity of each antigen was assessed by the geometric mean (GM) of expression determined by the Kaluza software.

Diagnoses of ETP-ALL and near-ETP-ALL used published criteria [1, 3, 4, 6, 15]. Patients not fulfilling the criteria for ETP-ALL or near-ETP-ALL were designated ‘conventional’ T-ALL (con-T-ALL). The intensity of CD5 expression on blasts was determined as the ratio between CD5-GM of T-lymphocytes within the sample to the CD5-GM of blasts (T-CD5: Bl-CD5 ratio) [15]. Our algorithm for classifying T-ALL patients into immunophenotypic subcategories is described in Supplementary Fig. 1A.

Flow cytometric MRD assessment

EOI-MRD was assessed in first-pull bone marrow aspiration (BMA) samples. The BMA samples were bulk-lysed with in-house prepared ammonium chloride-based lysis reagent and stained with an 11-antigen, 10-color cocktail (Supplementary Table 1). The processed samples were immediately fixed with 0.5% paraformaldehyde and acquired until the tube ran dry. The generated LMD files were analyzed using an in-house developed “mature antigen-based exclusion” approach adapted from Tembhare et al. [6]. Supplementary Fig. 1B details our sequential gating strategy for leukemia associated immunophenotype (LAIP) identification. A cluster of over 30 events with aberrant immunophenotype was considered for MRD quantification. The sensitivity of our MRD assay was 0.003% with a maximum coefficient of variation of 14.4% (refer to Supplementary Table 1 for our MRD assay validation and formula used for MRD calculation).

Antigen shift determination

Differences in expression intensity between baseline and EOI-residual blasts were analyzed for the following antigens (negative & positive controls): CD7 (B-lymphocytes & T-lymphocytes), CD4 (B-lymphocytes and CD4+ T-lymphocytes), CD8 (B-lymphocytes and CD8+ T-lymphocytes), CD5 (B-lymphocytes and T-lymphocytes), surface-CD3 (B-lymphocytes and T-lymphocytes), and CD38 (granulocytes and monocytes). Normalized mean fluorescence intensity (nMFI) for all these antigens was calculated for baseline and EOI-residual blasts as previously described [16].

For mature antigen-based MRD analysis, we analyzed the stability of mature T-cell associted antigens (CD7, CD4, CD8, CD5 and surface CD3) available in our MRD panel (Supplemental Table 1). CD38 was analyzed to assess the stability of this potentailly targetable antigen by daratumumab. Stability of CD56 could not be analyzed as both CD56 and CD16 were used in the BV510 fluorochrome of our MRD panel.

Statistical analyses

Statistical Package for Social Sciences (version 23, IBM, Armonk, NY) and MedCalc version 14.8.1 were used for statistical tests. For intergroup comparisons, Chi-squared and Mann-Whitney U tests were used. Occurrence of induction failure (≥5% BM blasts at EOI), relapse, and death were considered events. With the date of disease diagnosis as the starting time point, Kaplan–Meier survival analysis was used to determine 2-year rates of overall survival (OS), relapse-free survival (RFS), and event-free survival (EFS). Wilcoxon’s signed-rank test was used to assess differences in the expression intensity for CD4, CD8, CD5, CD7, CD38, and surface-CD3 (sCD3) antigens between leukemic blasts at diagnosis and residual blast at EOI-MRD. The risks incurred by the presence of mediastinal mass, hyperleukocytosis, immunophenotypic subtype of T-ALL, D8BNC status, and EOI-MRD positive status on OS, RFS, and EFS were determined by Cox proportional hazard model (Wald test). All statistical tests were two-tailed and considered significant at P≤0.05.

Among 306 consecutive treatment naïve ALL patients, 81 (36%) were of T-lineage origin. Of these 81 patients, the frequency of con-T-ALL, ETP-ALL and near-ETP-ALL was 60% (N=49), 21% (N=17) and 19% (N=15), respectively. Table 1 summarizes the clinico-laboratory characteristics of these patient categories.

Table 1 Clinical and laboratory characteristics of T-ALL subcategories.

ParametersOverall T-ALL (N=81)T-ALL subcategoriesP
Con-T-ALL
(N=49)
ETP-ALL
(N=17)
Near-ETP-ALL
(N=15)
ETP-ALL
vs.
Near-ETP-ALL
ETP-ALL
vs.
Con-T-ALL
Near-ETP-ALL
vs.
Con-T-ALL
Median (range) age in years17 (1–52)15 (1–50)17 (13–39)23 (5–52)0.8820.0030.016
Age group1.0000.0390.040
Pediatric (%)47 (58)34 (72%)7 (15%)6 (13%)
Adult (%)34 (42)15 (44%)10 (29%)9 (27%)
Sex (male:female)3.8:14.4:13.2:12.7:11.0000.6450.485
Median (range) Hb in g/L90 (30–142)90 (30–142)92 (30–131)88 (41–133)0.7371.0000.751
Median (range) WBC count, ×109/L64.1 (1–850)173 (1.1–850)70 (1–480)145 (3–590)0.0490.0050.751
Median (range) platelet, ×109/L54 (20–380)73 (20–366)125 (30–290)127 (20–380)0.9110.0080.080
Median (range) BM blast, %87 (22–99)87 (23–97)86 (22–98)89 (50–99)0.4730.7950.663
Median (range) PB blast, %78 (2–99)80 (2–97)42 (2–98)83 (2–99)0.1930.1740.411
Hyperleukocytosis41%45%18%53%0.0340.0460.567
Hepatomegaly42%42%27%58%0.2040.2830.319
Splenomegaly56%56%47%67%0.5160.5500.489
Lymphadenopathy78%73%87%86%1.0000.2900.342
Mediastinal mass31%36%33%13%0.3880.8420.095
CNS involvement at diagnosis3.2%2 (5)0%0%-0.4170.499
D8BNC35%32%54%20%0.2230.1680.440
EOI-MRD positive39% (N=59)22.5% (N=40)75%(N=12)71.4% (N=7)0.8650.0010.009
Relapse20% (N=60)18% (N=40)17% (N=12)38% (N=8)0.2920.9470.204
OS at 24 months65.2% (N=66)70.6% (N=42)60.4% (N=13)52% (N=11)0.1800.5510.019
RFS at 24 months76.1% (N=60)80% (N=40)79% (N=12)54.7% (N=8)0.2920.9560.190
EFS at 24 months64.5% (N=66)70.3% (N=42)66.6% (N=13)41% (N=11)0.0760.9780.013

Abbreviations: BM, bone marrow; CNS, central nervous system; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; Hb, hemoglobin; N, number of patients analyzed; NA, not applicable; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival; WBC, white blood cells.



Irrespective of immunophenotypic sub classification, T-ALL comprised 22% (47/209) and 35% (34/97) of our pediatric and adult ALL patients, respectively. T-ALL subtype specific clinico-laboratory profiles of our pediatric and adult T-ALL patients are presented in Table 2 and compared in Supplementary Table 2.

Table 2 Clinical and laboratory characteristics of immunophenotypic T-ALL subcategories among pediatric and adult age groups.

ParametersPediatric patientsAdult patients
T-ALL subtypePT-ALL subtypeP
Con-T-ALL
(N=34)
ETP-ALL
(N=7)
Near-ETP-ALL
(N=6)
ETP-ALL
vs.
Near- ETP-ALL
ETP-ALL
vs.
Con-T-ALL
Near- ETP-ALL
vs.
Con-T- T-ALL
Con-T-ALL
(N=15)
ETP-ALL
(N=10)
Near-ETP-ALL
(N=9)
ETP-ALL
vs.
Near- ETP-ALL
ETP-ALL
vs.
Con- T-ALL
Near- ETP-ALL
vs.
Con-T-ALL
Median age (range) in years12 (1–18)16 (13–17)13
(5–18)
0.2950.0560.34325
(20–50)
34
(19–39)
29
(20–52)
0.8420.3670.290
Sex (male:female)3.8:16:15:10.9060.3070.8256.5:12.3:12:10.8760.7020.243
Median (range) Hb in g/L91 (30–141)97
(30–131)
83
(41–129)
0.1810.4650.51789
(63–142)
80
(61–128)
88
(69–133)
0.3560.3381.000
Median (range) WBC, ×109/L110 (1.9–850)90.4
(3.2–267)
244
(3–590)
0.3660.1980.81088
(1.1–349)
55
(1–480)
68
(3.6–131)
0.0170.0360.682
Median (range) platelet, ×109/L83 (22–366)125
(30–245)
149
(32–380)
0.7310.1750.24052
(20–119)
125
(30–290)
100
(20–218)
0.7200.0160.138
Median (range) BM blast, %87 (23–97)86
(22–98)
95
(89–99)
0.1490.8450.02987
(64–96)
85
(38–95)
76
(50–97)
0.8630.7700.446
Median (range) PB blast, %84 (2–96)86
(2–98)
98
(2–99)
0.2680.8830.07461
(3–97)
36
(5–94)
76
(5–91)
0.1610.0730.770
Hyperleukocytosis53%29%67%0.0890.2400.53327%10%44%0.0890.3070.371
Hepatomegaly45%40%67%0.1090.8290.47836%20%56%0.1090.4040.349
Splenomegaly61.3%40%67&0.4630.3700.85543%50%67%0.4630.7290.265
Lymphadenopathy75%100%100%0.9060.2070.20770%80%78%0.9060.5600.658
Mediastinal mass40%20%17%0.1530.4020.28629%40%11%0.1530.5580.322
CNS involvement4%0%0%NA0.6380.6388%0%0%NA0.4520.620
Induction death7%0%0%0.0980.5080.5080%14%60%0.0980.1620.002
Induction failure0%0%0%NANANA0%17%75%0.0650.0010.001
D8BNC30%50%50%0.1050.3430.42240%57%0%0.1050.4860.074
EOI-MRD positive15%
(N=27)
83%
(N=6)
60%
(N=5)
0.3460.0010.02538.5%
(N=13)
67%
(N=6)
100%
(N=2)
0.3460.2520.104
Relapse11%
(N=27)
17%
(N=6)
17%
(N=6)
1.0000.7060.70631%
(N=13)
17%
(N=6)
100%
(N=2)
0.0350.2780.278
OS at 24 months79%
(N=29)
67%
(N=6)
100%
(N=6)
0.5640.8200.29748%
(N=13)
51%
(N=7)
0%
(N=5)
0.0250.5880.001
RFS at 24 months87%
(N=27)
83%
(N=6)
75%
(N=6)
0.9370.8050.72064%
(N=13)
75%
(N=6)
0%
(N=2)
0.0120.7050.014
EFS at 24 months81%
(N=29)
80%
(N=6)
75%
(N=6)
0.9370.8780.94345%
(N=13)
54%
(N=7)
0%
(N=5)
0.0190.767<0.001

Abbreviations: BM, bone marrow; CNS, the central nervous system; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; Hb, hemoglobin; N, number of patients analyzed; NA, not applicable; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival; WBC, white blood cells.



Antigen expression profile

FCM determined antigen expression profiles of all 81 T-ALL patients are presented in Supplementary Fig. 2. The median (range) T-CD5: Bl-CD5 expression ratio among con-T-ALL, near-ETP-ALL and ETP-ALL blasts was 1.83 (0.85–8.56), 3.39 (1.43–8.21) and 16.12 (11.06–59.21), respectively. Among con-T-ALL patients, 26.5% (N=13) had isolated CD4 expression, 10% (N=5) had isolated CD8 expression, dual expression for both CD4 and CD8 was observed in 45% (N=22) patients, and 20% (N=9) of the patients did not express either antigen. Expression frequency for myeloid/stem cell antigens (ETP-ALL vs. near-ETP-ALL patients) was CD117 (47% vs. 7%, P=0.011), CD34 (82% vs. 80%, P=0.865), HLA-DR (53% vs. 21%, P=0.073), CD13 (53% vs. 20%, P=0.055), CD33 (47% vs. 73%, P=0.131), and CD11b (29% vs. 21%, P=0.631).

Differences in the percentage of patients expressing immaturity associated antigens (CD10, CD34, and CD117), B-lineage antigens (CD19 and CD79a), myeloid antigens (CD13, CD11b, CD33), and non-lineage-specific antigens (CD123, CD56, and CD38) among our immunophenotypic subcategories of T-ALL are depicted in Fig. 1.

Fig. 1. Percentage of patients expressing lineage-specific and non-specific antigens across the immunophenotypic subtypes of T-ALL.

EOI-MRD assessment

Among 60 patients who completed induction, EOI-MRD was tested in 59 (40 con-T-ALL, 12 ETP-ALL, and 7 near-ETP-ALL). A median of 2.3 million events (range, 0.18 to 7.3 million) was acquired for analysis, and over 1.5 million events were acquired in 68% of the samples.

EOI-MRD was positive in 39% of the samples tested (32% of pediatric and 52% of adult samples). EOI-MRD was frequently positive among ETP-ALL (75%, P=0.001) and near-ETP-ALL (71%, P=0.009) patients, compared to con- T-ALL patients (22.5%).

Median (range) MRD quantified among con-T-ALL, ETP-ALL, and near-ETP-ALL samples was 0.192% (0.015–2.125), 5.360% (0.125–30.306), and 4.250% (0.532–10.436), respectively. There was a significant difference in EOI-MRD quantified between con-T-ALL vs. near-ETP-ALL patients (P=0.019), but not between con-T-ALL vs. ETP-ALL (P=0.074) and ETP-ALL vs. near-ETP-ALL (P=0.898) patients. Comparison of the clinico-laboratory profiles of our EOI-MRD positive and negative T-ALL patients, and their 2-year OS, RFS, and EFS rates are presented in Supplementary Table 3.

Age group-specific analysis revealed significantly different frequencies of EOI-MRD positivity between the subcategories of T-ALL (con-T-ALL vs. ETP-ALL vs. near-ETP- ALL) among pediatric (15% vs. 83.3% vs. 60%, P=0.002), but not adult T-ALL patients (38.5% vs. 67% vs. 100%, P=0.190).

Antigen stability

Analysis of the effect of induction therapy on expressions of CD4, CD8, CD7, CD5, CD38, and sCD3 antigens revealed statistically significant upregulations of CD8 (P=0.046) and CD38 (P=0.046) expression in EOI blasts of con-T-ALL and ETP-ALL patients, respectively (Supplementary Fig. 3).

OS, RFS, and EFS

Among the 81 T-ALL patients, 4 died before treatment and 11 left hospital care before initiating treatment (refer to Supplementary Fig. 4 for disease course during follow-up). Among the remaining 66 patients who were treated, 6 died during the induction phase (4.7% of con-T-ALL, 7.6% of ETP-ALL, and 27% of near-ETP-ALL; P=0.068). Induction failure was observed in 8.3% of ETP-ALL, 30% of near- ETP-ALL, and none of our con-T-ALL patients (P=0.005). A mean (±SD) follow-up of 12 (±10) months was available among 60 patients who had completed induction. None of the patients underwent hematopoietic stem cell transplantation.

Irrespective of the age at diagnosis and immunophenotypic subclassification, 2-year OS, RFS, and EFS rates among our T-ALL patients were 65%, 76%, and 64.5%, respectively. The survival profiles of our T-ALL patients pertinent to their immunophenotypic subcategorization are depicted in Table 1 and Fig. 2. Expression of CD56, CD19, and CD79a in the blasts did not have any significant impact (P>0.05) on the OS, RFS, and EFS among any of the immuno-phenotypic subcategories of T-ALL. The 2-year OS, RFS, and EFS specific to our pediatric and adult patients stratified by T-ALL subtypes are summarized in Table 2 and compared in Supplementary Table 2.

Fig. 2. Two-year overall survival (OS), relapse-free survival (RFS), and event-free survival (EFS) across all immunophenotypic subcategories of T-ALL analyzed together among all age groups (first row), pediatric patients (second row), and adult patients (third row).

Impact of EOI-MRD status on survival

Irrespective of immunophenotypic subclassification and age, there were significant differences in 2-year OS (86% vs. 48%, P=0.013), RFS (87% vs. 57%, P=0.022), and EFS (83% vs. 58%, P=0.008) between our EOI-MRD negative vs. positive T-ALL patients (refer to Supplementary Table 3 and Supplementary Fig. 5A for Kaplan–Meier survival curves).

Age group specific analysis revealed significant differences in 2-year OS (95% vs. 53%, P=0.044), RFS (95% vs. 60%, P=0.010), and EFS (95% vs. 60%, P=0.010) among EOI-MRD negative vs. positive pediatric T-ALL patients. However, 2-year OS (58% vs. 41%, P=0.303), RFS (67% vs. 54%, P=0.727) and EFS (50% vs. 44%, P=0.435) was not significantly different between EOI-MRD negative vs. positive adult T-ALL patients (refer to Supplementary Fig. 5B for Kaplan–Meier survival curves).

T-ALL subtype specific analysis revealed significant difference in 2-year OS (94% vs. 37%, P=0.012), RFS (94% vs. 50%, P=0.005), and EFS (94% vs. 50%, P=0.005) between EOI-MRD negative vs. positive pediatric con-T-ALL patients. However, there were no significant differences in 2-year OS (50% vs. 53%, P=0.874), RFS (62% vs. 66%, P=0.584) and EFS (42% vs. 53%, P=0.891) between the EOI-MRD negative vs. positive adult con-T-ALL patients (refer to Supplementary Fig. 5B for Kaplan–Meier survival curves). The lower number of patients available at the EOI timepoint in ETP-ALL (6 pediatric and 6 adult) and near-ETP-ALL (5 pediatric and 2) subtypes precluded analysis of age group specific impact of EOI-MRD status on survival in these categories.

Cox proportional hazard regression analysis was performed to identify risks incurred by the immunophenotypic subtype of T-ALL, presence of mediastinal mass and hyperleukocytosis at diagnosis, and D8BNC and EOI-MRD positive status on 2-year OS, RFS, and EFS on our pediatric and adult patients. The results are presented in Table 3.

Table 3 Univariate analysis of covariates with event-free, relapse-free, and overall survivals.

Variables2 years-EFS2 years-RFS2 years-OS
HR95% CIPHR95% CIPHR95% CIP
Pediatric univariateD8BNC0.6560.119–3.6200.6290.8160.135–4.9240.8250.3110.035–2.8010.298
Mediastinal mass1.8490.358–9.5490.4633.1090.347–27.8430.3111.5780.285–8.7210.601
Hyper-leucocytosis0.4220.082–2.1790.3030.6930.116–4.1540.6880.2280.027–1.9510.117
EOI-MRD positive10.1531.132–91.0960.03810.0811.123–90.4950.0397.3810.757–71.9520.085
Con T-ALL subtype1.1290.219–5.8210.8850.7010.117–4.1970.6982.2720.265–19.4900.454
ETP-ALL subtype0.8870.103–7.1320.8871.2270.137–10.9840.8550.9320.108–8.0140.949
Near-ETP-ALL subtype0.9560.115–7.9470.9671.4250.159–12.7570.7620.7800.091–6.7000.821
Adult univariateD8BNC1.1660.326–4.1720.8140.4480.046–4.3360.4881.4560.388–5.4620.577
Mediastinal mass3.0000.782–11.5020.1092.2100.426–11.4620.3115.0081.029–24.3740.056
Hyper-leucocytosis1.7840.615–5.1780.2874.0840.908–18.3680.0671.4820.483–4.5470.491
EOI-MRD positive1.6480.461–5.8830.4421.3020.291–5.8280.7302.0240.501–8.1850.323
Con-T-ALL subtype0.4250.144–1.2530.1210.6070.135–2.7380.5160.3610.117–1.1170.077
ETP-ALL subtype0.7300.203–2.6230.6300.4610.055–3.8330.4730.8240.226–3.0020.769
Near-ETP-ALL subtype7.9952.000–31.9680.00311.1221.533–80.7190.0176.6491.891–23.3830.003

Abbreviations: CI, confidence interval; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; HR, hazard ratio; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival.


Demography

The 15% frequency of ETP-ALL documented in our pediatric T-ALL patients is similar to previously observed frequencies of 11% and 14% [4, 6]. As reported in other studies [17, 18], we too observed adult age predilection for ETP-ALL (P=0.039) in our cohort. ETP-ALL compromised 29% of our adult T-ALL patients. The documented frequency is variable, with rates of 17% in the United States, 32% in Germany, and 47% in China [1, 18-22]. This marked heterogeneity might be due either to ethnic predisposition or incongruencies in FCI analysis, where both ETP-ALL and near-ETP-ALL are considered under a common ETP-ALL category.

The exact worldwide frequency of near-ETP-ALL is unknown, as only a few studies have recognized this entity [3, 4, 6, 19]. In the present study, near-ETP-ALL was also frequent among adult T-ALL patients (26%, P=0.040). This frequency is similar to the 33% frequency observed by Van Vlierberghe et al. [19]. The 13% frequency of near-ETP-ALL we observed is similar to the 17% frequency reported by the Children’s Oncology Group [4], but is higher than the 5.4% frequency reported by Tembhare et al. [6] from India.

Our results indicate that these immunophenotypic subcategories of T-ALL cannot be distinguished by the presence of hepatosplenomegaly, lymphadenopathy, or mediastinal mass at diagnosis (P>0.05). Regarding the laboratory parameters at diagnosis, our adult ETP-ALL patients had significantly lower white blood cell (P=0.036) and higher platelet (P=0.016) counts compared to those in our adult con- T-ALL patients. These findings are consistent with those of prior studies [21, 22]. However, Ma et al. [23] had observed significantly low white blood cell counts among their pediatric ETP-ALL patients.

Immunophenotype at diagnosis

Both ETP-ALL and near-ETP-ALL blasts are proposed to have originated from BM-derived early thymic precursor (ETP) cells that migrated to the thymus. These ETP cells are too immature and have a transcriptome profile enabling differentiation towards T, myeloid, and dendritic cell lineages [1]. The dendritic-lineage orientation of ETP-ALL and near-ETP-ALL blasts was reflected in our results, as we observed a high frequency of CD123 positivity (P<0.001) in these patients as compared to con-T-ALL patients (Fig. 1). In our T-ALL cohort, CD73 expression was restricted only to ETP-ALL and near-ETP-ALL blasts (P=0.007). CD86 was significantly expressed only among ETP-ALL blasts (P< 0.001) (Fig. 1). The diagnostic relevance of this observation has to be verified in a larger cohort.

Regarding cross-lineage antigen expression among T-ALL blasts, expression of the CD56 antigen of natural killer (NK) cells is frequently associated with ETP-ALL blasts and confers a poor prognosis [24-27]. Consistent with the literature, we also observed a higher frequency of CD56 expression in our ETP-ALL and near-ETP-ALL patients (P=0.009). However, in contrast to the available literature, expression of CD56 did not make any difference in the OS, EFS, and RFS of any immunophenotypic subcategories of our T-ALL patients (P>0.05). This contrast might reflect the smaller cohort size, differences in treatment protocols used, and limited follow-up available among our patients. Despite recent observations regarding the expression of B-lineage markers CD19 and CD79a in ETP-derived blasts [6, 28], the prognostic relevance of such aberrant expression is unknown. In the current study, CD19 expression was observed only among ETP-ALL and near-ETP-ALL blasts and not among con- T-ALL blasts (P=0.004). In contrast, CD79a expression was not predilected towards any of the immunophenotypic subcategories of T-ALL (P=0.172). Importantly, aberrant expression of either CD19 or CD79a did not translate into inferior 2-year survival outcomes in our con-T-ALL, ETP-ALL, and near-ETP-ALL patient categories (P>0.05).

EOI-MRD

Traditional T-ALL MRD assessment by FCM relies on identifying the expression of immaturity associated markers like CD34, TdT, and CD99 on CD7 and cytoplasmic CD3 expressing lymphocytes [4, 8, 9, 29]. This approach is not foolproof as these immaturity-related antigens are frequently down-regulated during treatment [29]. T-ALL MRD analysis by FCM is also hindered by the presence of NK cells and their precursors that can mimic residual disease [6]. Due to these shortcomings, most T-ALL MRD data are by high-throughput sequencing for IgG and TCR rearrangements. The literature on T-MRD by FCM is limited [4, 8, 9, 29, 30]. However, these molecular MRD detection techniques might not be successful in ETP-ALL and near-ETP-ALL samples as these leukemias originate from precursor cells that are too immature to have undergone TCR rearrangement [3].

With the increased availability of ≥8 color flow cytometers, the results and sensitivity of T-MRD assessment by FCM are highly comparable to molecular T-MRD assays [8]. Use of 8–9 color panels by the Children’s Oncology Group (COG) yielded EOI-MRD detection rates of 30.5%, 81.4%, and 64.8% in pediatric (N=1,144) con-T-ALL, ETP-ALL, and near-ETP-ALL patients, respectively [4]. In an Indian study in which 35 T-ALL patients of all age groups were analyzed using an 8-color panel, EOI-MRD was detectable in 37% of the patients [30]. A recent study discussed the experience with T-ALL MRD using an 11-antigen 10-color FCM panel. Use of a mature T-cell antigen-based “exclusion” approach for gating detected EOI-MRD in 46.5% of the pediatric T-ALL cohort (N=269) [6]. In all these studies, cumulative MRD positivity observed among ETP-ALL and near-ETP-ALL patients was higher than the MRD positivity observed among con-T-ALL patients (74% with P<0.001 [6], 73.1% by the COG [4], and 67% with P=0.033 [30]). Our result was similar (73% with P<0.001).

Tembhare et al. [6] observed subtle down-regulation of surface CD3, CD4, CD5, CD8, and CD38 expression in their EOI-residual blasts. In the current study, we observed stable expression of CD4, CD5, CD7, and surface CD3 between the baseline and EOI-residual leukemic blasts (P>0.05) among all immunophenotypic subcategories of T-ALL. However, there was upregulated expression of CD8 (P=0.046) and CD38 (P=0.046) in the EOI blasts of con-T-ALL and ETP-ALL samples, respectively (Supplementary Fig. 3). Tembhare et al. [6] described that these immunophenotype shifts were too subtle to interfere with MRD analysis. Our findings are similar. These collective results indicate that the mature T-cell-related antigen-based approach is reliable for T-MRD analysis by FCM. Importantly, the expression of CD38 in nearly 98% of T-ALL patients at diagnosis in the present and previous [6] studies, along with the stable/upregulated expression of this antigen in the EOI-residual blasts, can be used as a potential target for daratumumab therapy [31]. In our cohort, CD73 expression was specifically associated with ETP-ALL and near-ETP-ALL patients (Fig. 2). Recent optimistic results were obtained with the use of anti-CD73 monoclonal antibody-based treatment in solid tumors [32]. Validation of the stability and specific expression of CD73 in ETP-ALL and near-ETP-ALL patients in a larger cohort would provide a rationale for initiating anti- CD73-based clinical trials in the treatment of these patients.

EOI-MRD status among our ETP-ALL and near-ETP-ALL patients was independent of their baseline clinical and laboratory characteristics. However, our EOI-MRD negative con-T-ALL patients were frequently diagnosed with mediastinal mass, high white blood cell count, and hyperleucocytosis at diagnosis as compared to those in the EOI-MRD positive counterparts.

Outcome in pediatric patients

Conter et al. [33] observed D8BNC status among 55% of their pediatric ETP-ALL patients. This result is similar to that in our cohort, where 50% of patients in each of the ETP-ALL and near-ETP-ALL categories were steroid unresponsive on day 8 (Table 2). High incidence of induction failure and disease relapse has been documented among pediatric ETP-ALL patients [2, 4, 23, 34]. However, none of our pediatric con-T-ALL, ETP-ALL, or near-ETP-ALL patients experienced induction failure or had a significantly higher relapse (Table 2). These contrasting observation have to be validated in a larger cohort with extended follow-up.

We observed no differences (P>0.05) in the 2-year OS, RFS, and EFS among our pediatric con-T-ALL (79%, 83%, and 81%, respectively), ETP-ALL (67%, 87%, and 80%), and near-ETP-ALL (100%, 75%, and 75%) patients. Consistently, there were no significant differences in 5-year OS and EFS among the con-T (92% and 86.9%, respectively), ETP-ALL (93% and 87%), and near-ETP-ALL (91.6% and 84.4%) patients documented in the largest ever pediatric T-ALL data by the Children’s Oncology Group [4]. However, inferior 2-, 4-, 5-, and 10-year OS and EFS have also been documented among pediatric-ETP-ALL patients from Italy, Japan, China, and USA, respectively [2, 23, 34]. These different observations might be due to heterogeneity in the sample sizes and ethnicities of the study cohorts. Hence, the true prognostic relevance of both ETP-ALL and near-ETP-ALL will be determined only by prospective studies with a large sample size and longer follow-up.

Irrespective of immunophenotypic sub classification, our EOI-MRD negative pediatric T-ALL patients had significantly better 2-year OS (95% vs. 53%, P=0.044), RFS (95% vs. 60%, P=0.010), and EFS (95% vs. 72%, P=0.010) than their EOI-MRD positive counterparts, consistent with previous observations [7].

Outcome in adult patients

In the current study, we did not observe any differences in 2-year OS, EFS, and RFS between our adult con-T-ALL and ETP-ALL patients (Table 2). These findings are consistent with two prior studies [21, 22], but discordant with the inferior OS documented among adult & adolescent ETP-ALL patients by Jain et al. [20]. Importantly, our adult near- ETP-ALL patients had the most inferior 2-year OS, EFS, and RFS as compared to that in the con-T-ALL and ETP-ALL patients diagnosed in this age group (Table 2).

An important observation from our study is the EOI-MRD positive status (irrespective of T-ALL subtype) and the presence of near-ETP-ALL immunophenotype (irrespective of EOI-MRD status) influencing the 2-year survival profile among our pediatric and adult T-ALL patients, respectively. These results are encouraging concerning hematopoietic stem cell transplantation at first remission in EOI-MRD positive pediatric T-ALL patients and all adult T-ALL patients diagnosed with near-ETP-ALL. However, the small age-specific sample size in our study precludes any definite conclusions.

Both ETP-ALL and near-ETP-ALL are common among adult T-ALL patients. Irrespective of age at diagnosis, both these entities are associated with a high frequency of EOI-MRD positivity. Our results indicate adverse 2-year survival conferred by the presence of EOI-MRD positivity among pediatric T-ALL patients and by the diagnosis of near-ETP-ALL phenotype among adult T-ALL patients. However, large prospective clinical trials are warranted to confirm these conclusions.

Study limitations

Being a relatively rare disease, the number of patients in our cohort might not be powered for exact outcome analysis across each immunophenotypic subcategory of T-ALL. Hence, the survival outcomes documented in our study have to be validated in a larger cohort. The mutational profile of leukemic lymphoblasts was not evaluated in our patients. Being retrospective data, our study results are purely observational and did not explore the underlying leukemogenic differences between the subtypes of T-ALL.

Mrs. Arcot Radhakrishnan Abitha (Senior scientific assistant) is acknowledged for her efforts in FCM sample processing and sample acquisition. Dr. Rama R, assistant professor and statistician, helped with the statistics involved in the manuscript.

No potential conflicts of interest relevant to this article were reported.

  1. Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. Revised 4th ed. Lyon, France:. IARC Press,:421.
  2. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol 2009;10:147-56.
    Pubmed KoreaMed CrossRef
  3. Wu D, Sherwood A, Fromm JR, et al. High-throughput sequencing detects minimal residual disease in acute T lymphoblastic leukemia. Sci Transl Med 2012;4:134ra63.
    Pubmed CrossRef
  4. Wood BL, Winter SS, Dunsmore KP, et al. T-lymphoblastic leukemia (T-ALL) shows excellent outcome, lack of significance of the early thymic precursor (ETP) immunophenotype, and validation of the prognostic value of end-induction minimal residual disease (MRD) in Children's Oncology Group (COG) Study AALL0434. Blood (ASH Annual Meeting Abstracts) 2014;124:1.
    CrossRef
  5. Lin N, Liu ZH, Wang PP, Fu W, Yan XJ, Li Y. Immunophenotypic analysis of patients with adult acute T-lymphoblastic leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2020;28:442-5.
    Pubmed CrossRef
  6. Tembhare PR, Chatterjee G, Khanka T, et al. Eleven-marker 10-color flow cytometric assessment of measurable residual disease for T-cell acute lymphoblastic leukemia using an approach of exclusion. Cytometry B Clin Cytom 2021;100:421-33.
    Pubmed CrossRef
  7. Tembhare PR, Narula G, Khanka T, et al. Post-induction measurable residual disease using multicolor flow cytometry is strongly predictive of inferior clinical outcome in the real-life management of childhood T-cell acute lymphoblastic leukemia: a study of 256 patients. Front Oncol 2020;10:577.
    Pubmed KoreaMed CrossRef
  8. Modvig S, Madsen HO, Siitonen SM, et al. Minimal residual disease quantification by flow cytometry provides reliable risk stratification in T-cell acute lymphoblastic leukemia. Leukemia 2019;33:1324-36.
    Pubmed CrossRef
  9. Thörn I, Forestier E, Botling J, et al. Minimal residual disease assessment in childhood acute lymphoblastic leukaemia: a Swedish multi‐centre study comparing real‐time polymerase chain reaction and multicolour flow cytometry. Br J Haematol 2011;152:743-53.
    Pubmed CrossRef
  10. Gudapati P, Khanka T, Chatterjee G, et al. CD304/neuropilin‐1 is a very useful and dependable marker for the measurable residual disease assessment of B‐cell precursor acute lymphoblastic leukemia. Cytometry B Clin Cytom 2020;98:328-35.
    Pubmed CrossRef
  11. Manabe A, Ohara A, Hasegawa D, et al. Significance of the complete clearance of peripheral blasts after 7 days of prednisolone treatment in children with acute lymphoblastic leukemia: the Tokyo Children's Cancer Study Group Study L99-15. Haematologica 2008;93:1155-60.
    Pubmed CrossRef
  12. Cherian T, John R, Joseph LL, et al. Complete peripheral blast clearance is superior to the conventional cut-off of 1000/µL in predicting relapse in pediatric pre-B acute lymphoblastic leukemia. Indian J Hematol Blood Transfus 2021;37:366-71.
    Pubmed KoreaMed CrossRef
  13. Bommannan BKK, Arumugam JR, Sundersingh S, Rajan PT, Radhakrishnan V, Sagar TG. CD19 negative and dim precursor B-lineage acute lymphoblastic leukemias: real-world challenges in a targeted-immunotherapy era. Leuk Lymphoma 2019;60:3154-60.
    Pubmed CrossRef
  14. Dworzak MN, Buldini B, Gaipa G, et al. AIEOP‐BFM consensus guidelines 2016 for flow cytometric immunophenotyping of pediatric acute lymphoblastic leukemia. Cytometry B Clin Cytom 2018;94:82-93.
    Pubmed CrossRef
  15. Chopra A, Bakhshi S, Pramanik SK, et al. Immunophenotypic analysis of T‐acute lymphoblastic leukemia. A CD 5‐based ETP‐ALL perspective of non‐ETP T‐ALL. Eur J Haematol 2014;92:211-8.
    Pubmed CrossRef
  16. Sędek Ł, Theunissen P, Sobral da Costa E, et al. Differential expression of CD73, CD86 and CD304 in normal vs. leukemic B-cell precursors and their utility as stable minimal residual disease markers in childhood B-cell precursor acute lymphoblastic leukemia. J Immunol Methods 2019;475:112429.
    Pubmed CrossRef
  17. Haydu JE, Ferrando AA. Early T-cell precursor acute lymphoblastic leukaemia. Curr Opin Hematol 2013;20:369-73.
    Pubmed KoreaMed CrossRef
  18. Neumann M, Heesch S, Gökbuget N, et al. Clinical and molecular characterization of early T-cell precursor leukemia: a high-risk subgroup in adult T-ALL with a high frequency of FLT3 mutations. Blood Cancer J 2012;2:e55.
    Pubmed KoreaMed CrossRef
  19. Van Vlierberghe P, Ambesi-Impiombato A, Perez-Garcia A, et al. ETV6 mutations in early immature human T cell leukemias. J Exp Med 2011;208:2571-9.
    Pubmed KoreaMed CrossRef
  20. Jain N, Lamb AV, O'Brien S, et al. Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype. Blood 2016;127:1863-9.
    Pubmed KoreaMed CrossRef
  21. Zhang Y, Qian JJ, Zhou YL, et al. Comparison of early T-cell precursor and non-ETP subtypes among 122 Chinese adults with acute lymphoblastic leukemia. Front Oncol 2020;10:1423.
    Pubmed KoreaMed CrossRef
  22. Liao HY, Sun ZY, Wang YX, Jin YM, Zhu HL, Jiang NG. Outcome of 126 adolescent and adult T-cell acute leukemia/lymphoma patients and the prognostic significance of early T-cell precursor leukemia subtype. Zhonghua Xue Ye Xue Za Zhi 2019;40:561-7.
    Pubmed KoreaMed CrossRef
  23. Ma M, Wang X, Tang J, et al. Early T-cell precursor leukemia: a subtype of high risk childhood acute lymphoblastic leukemia. Front Med 2012;6:416-20.
    Pubmed CrossRef
  24. Montero I, Rios E, Parody R, Perez-Hurtado JM, Martin-Noya A, Rodriguez JM. CD56 in T-cell acute lymphoblastic leukaemia: a malignant transformation of an early myeloid-lymphoid progenitor? Haematologica 2003;88:ELT26.
    Pubmed
  25. Fischer L, Gökbuget N, Schwartz S, et al. CD56 expression in T-cell acute lymphoblastic leukemia is associated with non-thymic phenotype and resistance to induction therapy but no inferior survival after risk-adapted therapy. Haematologica 2009;94:224-9.
    Pubmed KoreaMed CrossRef
  26. Fuhrmann S, Schabath R, Möricke A, et al. Expression of CD56 defines a distinct subgroup in childhood T‐ALL with inferior outcome. Results of the ALL‐BFM 2000 trial. Br J Haematol 2018;183:96-103.
    Pubmed CrossRef
  27. Dalmazzo LF, Jácomo RH, Marinato AF, et al. The presence of CD56/CD16 in T‐cell acute lymphoblastic leukaemia correlates with the expression of cytotoxic molecules and is associated with worse response to treatment. Br J Haematol 2009;144:223-9.
    Pubmed CrossRef
  28. Garg S, Gupta SK, Bakhshi S, Mallick S, Kumar L. ETP-ALL with aberrant B marker expression: case series and a brief review of literature. Int J Lab Hematol 2019;41:e32-7.
    Pubmed CrossRef
  29. Roshal M, Fromm JR, Winter S, Dunsmore K, Wood BL. Immaturity associated antigens are lost during induction for T cell lymphoblastic leukemia: implications for minimal residual disease detection. Cytometry B Clin Cytom 2010;78:139-46.
    Pubmed KoreaMed CrossRef
  30. Singh N, Agrawal N, Sood R, et al. T-ALL minimal residual disease using a simplified gating strategy and its clinico-hematologic correlation: a single center experience from North India. Indian J Hematol Blood Transfus 2019;35:707-10.
    Pubmed KoreaMed CrossRef
  31. Tembhare PR, Sriram H, Khanka T, et al. Flow cytometric evaluation of CD38 expression levels in the newly diagnosed T-cell acute lymphoblastic leukemia and the effect of chemotherapy on its expression in measurable residual disease, refractory disease and relapsed disease: an implication for anti-CD38 immunotherapy. J Immunother Cancer 2020;8:e000630.
    Pubmed KoreaMed CrossRef
  32. Chen S, Wainwright DA, Wu JD, et al. CD73: an emerging checkpoint for cancer immunotherapy. Immunotherapy 2019;11:983-97.
    Pubmed KoreaMed CrossRef
  33. Conter V, Valsecchi MG, Buldini B, et al. Early T-cell precursor acute lymphoblastic leukaemia in children treated in AIEOP centres with AIEOP-BFM protocols: a retrospective analysis. Lancet Haematol 2016;3:e80-6.
    Pubmed CrossRef
  34. Inukai T, Kiyokawa N, Campana D, et al. Clinical significance of early T‐cell precursor acute lymphoblastic leukaemia: results of the Tokyo Children's Cancer Study Group Study L99‐15. Br J Haematol 2012;156:358-65.
    Pubmed CrossRef

Article

Original Article

Blood Res 2022; 57(3): 175-196

Published online September 30, 2022 https://doi.org/10.5045/br.2022.2022104

Copyright © The Korean Society of Hematology.

Relevance of flow cytometric categorization and end-of-induction measurable residual disease assessment in pediatric and adult T-lymphoblastic leukemia patients

Karthik Bommannan1, Jhansi Rani Arumugam1, Venkatraman Radhakrishnan2, Jayachandran Perumal Kalaiyarasi2, Parathan Karunakaran2, Nikita Mehra2, Tenali Gnana Sagar2, Shirley Sundersingh1

Departments of 1Oncopathology and 2Medical Oncology, Cancer Institute (W.I.A.), Adyar, India

Correspondence to:Karthik Bommannan, M.D., D.M.
Department of Oncopathology, Cancer Institute (W.I.A), Adyar, Chennai 600020, India
E-mail: bkkb87@gmail.com

*This study was supported by a grant from Indian Council of Medical Research (ICMR) and the Indian Childhood Collaborative Leukemia (ICiCLe) project of the National Cancer Grid (NCG).

Received: May 16, 2022; Revised: June 15, 2022; Accepted: June 21, 2022

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Background
T-lymphoblastic leukemia (T-ALL) patients expressing myeloid/stem cell antigens are classified as early T-cell precursor lymphoblastic leukemia (ETP-ALL) or near-ETP-ALL.
Methods
Clinico-laboratory profiles, flow cytometric end-of-induction measurable residual disease (EOI-MRD), and survival of treatment naïve T-ALL patients were analyzed according to their immunophenotypic subtypes.
Results
Among 81 consecutive T-ALL patients diagnosed, 21% (N=17) were ETP-ALL and 19% (N=15) were near-ETP-ALL. EOI-MRD was detectable in 39% of the 59 samples tested (31.6% of pediatric samples and 52.4% of adult samples). The frequency of EOI-MRD positivity was significantly higher among ETP-ALL (75%, P=0.001) and near-ETP-ALL (71%, P=0.009) patients compared to that in conventional-T-ALL (con-T-ALL) patients (22.5%). CD8 (P=0.046) and CD38 (P=0.046) expressions were significantly upregulated in the EOI blasts of con-T-ALL and ETP-ALL samples, respectively. The 2-year rates of overall (OS), relapse-free (RFS), and event-free survival (EFS) among the T-ALL patients (pediatric vs. adult) was 79.5% vs. 39.8% (P<0.001), 84.3% vs. 60.4% (P=0.026), and 80.3% vs. 38% (P<0.001), respectively. Univariate analysis revealed that 2-year EFS and RFS of pediatric T-ALL patients was independent of T-ALL subtype and was influenced only by EOI-MRD status. However, 2-year OS, RFS, and EFS among adult T-ALL patients were EOI-MRD independent and influenced only by the near-ETP-ALL phenotype.
Conclusion
Two-year survival among pediatric and adult T-ALL patients is attributed to EOI-MRD status and near-ETP-ALL phenotype, respectively.

Keywords: Measurable residual disease, Flow cytometry, T-lineage acute lymphoblastic leuke mia, ETP-ALL, Near-ETP-ALL

INTRODUCTION

T-lymphoblastic leukemia (T-ALL) comprises 15% of pediatric and 25% of adult acute lymphoblastic leukemia patients [1]. First described by Coustan-Smith et al. [2] in 2009, ‘early T-cell precursor lymphoblastic leukemia’ (ETP-ALL) is a subtype of T-ALL in which T-lymphoblasts express myeloid/stem cell-associated antigens in the absence of CD1a, CD5, and CD8 expression. ‘Near-ETP-ALL’ is a T-ALL subtype recognized by the World Health Organization (WHO) in 2017. In this subtype, T-lymphoblasts meet all immunophenotype criteria for ETP-ALL, except for having significant CD5 expression [1-3]. Clinical and laboratory characteristics of pediatric and adult ETP-ALL patients have been documented. However, data of near-ETP-ALL patients are limited [4, 5]. Also, the role of other lineage-specific and non-lineage-specific antigens that could distinguish between T-ALL subtypes is unknown.

Flow cytometric measurable residual disease (FCM-MRD) assessment is important for the risk-adapted management of B-lymphoblastic leukemia (B-ALL) patients. However, FCM-MRD-based treatment decisions are not yet part of the management protocols for T-ALL patients. This reflects the limited availability of literature on FCM-MRD in T-ALL. Most of the available publications have included both ETP-ALL and near-ETP-ALL as a common category for data analysis [4, 6-9].

To the best of our knowledge, data comparing age group specific clinico-laboratory profiles across the immunophenotypic subcategories of T-ALL patients are still lacking. Presently, we share our experience regarding clinico-laboratory profiles, end-of-induction (EOI) FCM-MRD, and 2-year survival outcomes of pediatric and adult T-ALL patients immunophenotypically subclassified according to the WHO 2017 guidelines.

MATERIALS AND METHODS

This retrospective study was approved by our Institute’s ethics committee. All treatment naïve T-ALL patients diagnosed between December 2017 to March 2020 were included. T-ALL was diagnosed by morphologic evaluation of peripheral blood (PB) and bone marrow (BM) aspiration smears, followed by a 10-color FCM analysis (Supplementary Table 1). Hyperleukocytosis was defined as ≥100×109/L leukocytes in PB [7]. Pediatric (age ≤18 yr) and adult patients were treated with the Indian Collaborative Childhood Leukemia group (high risk-arm) and Berlin-Frankfurt-Muenster (BFM) 95 protocols, respectively [10]. Treatment protocols were not influenced by T-ALL immunophenotype subcategory or end-of-induction measurable residual disease (EOI-MRD) status. During induction, an absolute PB blast count ≥1,000 cells/µL on day 8 of treatment was considered ‘day 8 blasts not cleared’ (D8BNC) status [11, 12].

Diagnostic flow cytometry

BM samples were processed using our previously described ‘lyse-stain-wash’ protocol [13]. A minimum of 100,000 events were acquired per tube using a Beckman Coulter Navios EX flow cytometer. Generated list-mode data (LMD) files were analyzed with Kaluza (Version 2.0) software (Beckman Coulter) using our in-house developed analysis templates. The antigen expression profile was reported according to the Associazione Italiana Ematologia Oncologia Pediatrica-BFM (AIEOP-BFM) 2016 recommendations [14]. The expression intensity of each antigen was assessed by the geometric mean (GM) of expression determined by the Kaluza software.

Diagnoses of ETP-ALL and near-ETP-ALL used published criteria [1, 3, 4, 6, 15]. Patients not fulfilling the criteria for ETP-ALL or near-ETP-ALL were designated ‘conventional’ T-ALL (con-T-ALL). The intensity of CD5 expression on blasts was determined as the ratio between CD5-GM of T-lymphocytes within the sample to the CD5-GM of blasts (T-CD5: Bl-CD5 ratio) [15]. Our algorithm for classifying T-ALL patients into immunophenotypic subcategories is described in Supplementary Fig. 1A.

Flow cytometric MRD assessment

EOI-MRD was assessed in first-pull bone marrow aspiration (BMA) samples. The BMA samples were bulk-lysed with in-house prepared ammonium chloride-based lysis reagent and stained with an 11-antigen, 10-color cocktail (Supplementary Table 1). The processed samples were immediately fixed with 0.5% paraformaldehyde and acquired until the tube ran dry. The generated LMD files were analyzed using an in-house developed “mature antigen-based exclusion” approach adapted from Tembhare et al. [6]. Supplementary Fig. 1B details our sequential gating strategy for leukemia associated immunophenotype (LAIP) identification. A cluster of over 30 events with aberrant immunophenotype was considered for MRD quantification. The sensitivity of our MRD assay was 0.003% with a maximum coefficient of variation of 14.4% (refer to Supplementary Table 1 for our MRD assay validation and formula used for MRD calculation).

Antigen shift determination

Differences in expression intensity between baseline and EOI-residual blasts were analyzed for the following antigens (negative & positive controls): CD7 (B-lymphocytes & T-lymphocytes), CD4 (B-lymphocytes and CD4+ T-lymphocytes), CD8 (B-lymphocytes and CD8+ T-lymphocytes), CD5 (B-lymphocytes and T-lymphocytes), surface-CD3 (B-lymphocytes and T-lymphocytes), and CD38 (granulocytes and monocytes). Normalized mean fluorescence intensity (nMFI) for all these antigens was calculated for baseline and EOI-residual blasts as previously described [16].

For mature antigen-based MRD analysis, we analyzed the stability of mature T-cell associted antigens (CD7, CD4, CD8, CD5 and surface CD3) available in our MRD panel (Supplemental Table 1). CD38 was analyzed to assess the stability of this potentailly targetable antigen by daratumumab. Stability of CD56 could not be analyzed as both CD56 and CD16 were used in the BV510 fluorochrome of our MRD panel.

Statistical analyses

Statistical Package for Social Sciences (version 23, IBM, Armonk, NY) and MedCalc version 14.8.1 were used for statistical tests. For intergroup comparisons, Chi-squared and Mann-Whitney U tests were used. Occurrence of induction failure (≥5% BM blasts at EOI), relapse, and death were considered events. With the date of disease diagnosis as the starting time point, Kaplan–Meier survival analysis was used to determine 2-year rates of overall survival (OS), relapse-free survival (RFS), and event-free survival (EFS). Wilcoxon’s signed-rank test was used to assess differences in the expression intensity for CD4, CD8, CD5, CD7, CD38, and surface-CD3 (sCD3) antigens between leukemic blasts at diagnosis and residual blast at EOI-MRD. The risks incurred by the presence of mediastinal mass, hyperleukocytosis, immunophenotypic subtype of T-ALL, D8BNC status, and EOI-MRD positive status on OS, RFS, and EFS were determined by Cox proportional hazard model (Wald test). All statistical tests were two-tailed and considered significant at P≤0.05.

RESULTS

Among 306 consecutive treatment naïve ALL patients, 81 (36%) were of T-lineage origin. Of these 81 patients, the frequency of con-T-ALL, ETP-ALL and near-ETP-ALL was 60% (N=49), 21% (N=17) and 19% (N=15), respectively. Table 1 summarizes the clinico-laboratory characteristics of these patient categories.

Table 1 . Clinical and laboratory characteristics of T-ALL subcategories..

ParametersOverall T-ALL (N=81)T-ALL subcategoriesP
Con-T-ALL
(N=49)
ETP-ALL
(N=17)
Near-ETP-ALL
(N=15)
ETP-ALL
vs.
Near-ETP-ALL
ETP-ALL
vs.
Con-T-ALL
Near-ETP-ALL
vs.
Con-T-ALL
Median (range) age in years17 (1–52)15 (1–50)17 (13–39)23 (5–52)0.8820.0030.016
Age group1.0000.0390.040
Pediatric (%)47 (58)34 (72%)7 (15%)6 (13%)
Adult (%)34 (42)15 (44%)10 (29%)9 (27%)
Sex (male:female)3.8:14.4:13.2:12.7:11.0000.6450.485
Median (range) Hb in g/L90 (30–142)90 (30–142)92 (30–131)88 (41–133)0.7371.0000.751
Median (range) WBC count, ×109/L64.1 (1–850)173 (1.1–850)70 (1–480)145 (3–590)0.0490.0050.751
Median (range) platelet, ×109/L54 (20–380)73 (20–366)125 (30–290)127 (20–380)0.9110.0080.080
Median (range) BM blast, %87 (22–99)87 (23–97)86 (22–98)89 (50–99)0.4730.7950.663
Median (range) PB blast, %78 (2–99)80 (2–97)42 (2–98)83 (2–99)0.1930.1740.411
Hyperleukocytosis41%45%18%53%0.0340.0460.567
Hepatomegaly42%42%27%58%0.2040.2830.319
Splenomegaly56%56%47%67%0.5160.5500.489
Lymphadenopathy78%73%87%86%1.0000.2900.342
Mediastinal mass31%36%33%13%0.3880.8420.095
CNS involvement at diagnosis3.2%2 (5)0%0%-0.4170.499
D8BNC35%32%54%20%0.2230.1680.440
EOI-MRD positive39% (N=59)22.5% (N=40)75%(N=12)71.4% (N=7)0.8650.0010.009
Relapse20% (N=60)18% (N=40)17% (N=12)38% (N=8)0.2920.9470.204
OS at 24 months65.2% (N=66)70.6% (N=42)60.4% (N=13)52% (N=11)0.1800.5510.019
RFS at 24 months76.1% (N=60)80% (N=40)79% (N=12)54.7% (N=8)0.2920.9560.190
EFS at 24 months64.5% (N=66)70.3% (N=42)66.6% (N=13)41% (N=11)0.0760.9780.013

Abbreviations: BM, bone marrow; CNS, central nervous system; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; Hb, hemoglobin; N, number of patients analyzed; NA, not applicable; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival; WBC, white blood cells..



Irrespective of immunophenotypic sub classification, T-ALL comprised 22% (47/209) and 35% (34/97) of our pediatric and adult ALL patients, respectively. T-ALL subtype specific clinico-laboratory profiles of our pediatric and adult T-ALL patients are presented in Table 2 and compared in Supplementary Table 2.

Table 2 . Clinical and laboratory characteristics of immunophenotypic T-ALL subcategories among pediatric and adult age groups..

ParametersPediatric patientsAdult patients
T-ALL subtypePT-ALL subtypeP
Con-T-ALL
(N=34)
ETP-ALL
(N=7)
Near-ETP-ALL
(N=6)
ETP-ALL
vs.
Near- ETP-ALL
ETP-ALL
vs.
Con-T-ALL
Near- ETP-ALL
vs.
Con-T- T-ALL
Con-T-ALL
(N=15)
ETP-ALL
(N=10)
Near-ETP-ALL
(N=9)
ETP-ALL
vs.
Near- ETP-ALL
ETP-ALL
vs.
Con- T-ALL
Near- ETP-ALL
vs.
Con-T-ALL
Median age (range) in years12 (1–18)16 (13–17)13
(5–18)
0.2950.0560.34325
(20–50)
34
(19–39)
29
(20–52)
0.8420.3670.290
Sex (male:female)3.8:16:15:10.9060.3070.8256.5:12.3:12:10.8760.7020.243
Median (range) Hb in g/L91 (30–141)97
(30–131)
83
(41–129)
0.1810.4650.51789
(63–142)
80
(61–128)
88
(69–133)
0.3560.3381.000
Median (range) WBC, ×109/L110 (1.9–850)90.4
(3.2–267)
244
(3–590)
0.3660.1980.81088
(1.1–349)
55
(1–480)
68
(3.6–131)
0.0170.0360.682
Median (range) platelet, ×109/L83 (22–366)125
(30–245)
149
(32–380)
0.7310.1750.24052
(20–119)
125
(30–290)
100
(20–218)
0.7200.0160.138
Median (range) BM blast, %87 (23–97)86
(22–98)
95
(89–99)
0.1490.8450.02987
(64–96)
85
(38–95)
76
(50–97)
0.8630.7700.446
Median (range) PB blast, %84 (2–96)86
(2–98)
98
(2–99)
0.2680.8830.07461
(3–97)
36
(5–94)
76
(5–91)
0.1610.0730.770
Hyperleukocytosis53%29%67%0.0890.2400.53327%10%44%0.0890.3070.371
Hepatomegaly45%40%67%0.1090.8290.47836%20%56%0.1090.4040.349
Splenomegaly61.3%40%67&0.4630.3700.85543%50%67%0.4630.7290.265
Lymphadenopathy75%100%100%0.9060.2070.20770%80%78%0.9060.5600.658
Mediastinal mass40%20%17%0.1530.4020.28629%40%11%0.1530.5580.322
CNS involvement4%0%0%NA0.6380.6388%0%0%NA0.4520.620
Induction death7%0%0%0.0980.5080.5080%14%60%0.0980.1620.002
Induction failure0%0%0%NANANA0%17%75%0.0650.0010.001
D8BNC30%50%50%0.1050.3430.42240%57%0%0.1050.4860.074
EOI-MRD positive15%
(N=27)
83%
(N=6)
60%
(N=5)
0.3460.0010.02538.5%
(N=13)
67%
(N=6)
100%
(N=2)
0.3460.2520.104
Relapse11%
(N=27)
17%
(N=6)
17%
(N=6)
1.0000.7060.70631%
(N=13)
17%
(N=6)
100%
(N=2)
0.0350.2780.278
OS at 24 months79%
(N=29)
67%
(N=6)
100%
(N=6)
0.5640.8200.29748%
(N=13)
51%
(N=7)
0%
(N=5)
0.0250.5880.001
RFS at 24 months87%
(N=27)
83%
(N=6)
75%
(N=6)
0.9370.8050.72064%
(N=13)
75%
(N=6)
0%
(N=2)
0.0120.7050.014
EFS at 24 months81%
(N=29)
80%
(N=6)
75%
(N=6)
0.9370.8780.94345%
(N=13)
54%
(N=7)
0%
(N=5)
0.0190.767<0.001

Abbreviations: BM, bone marrow; CNS, the central nervous system; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; Hb, hemoglobin; N, number of patients analyzed; NA, not applicable; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival; WBC, white blood cells..



Antigen expression profile

FCM determined antigen expression profiles of all 81 T-ALL patients are presented in Supplementary Fig. 2. The median (range) T-CD5: Bl-CD5 expression ratio among con-T-ALL, near-ETP-ALL and ETP-ALL blasts was 1.83 (0.85–8.56), 3.39 (1.43–8.21) and 16.12 (11.06–59.21), respectively. Among con-T-ALL patients, 26.5% (N=13) had isolated CD4 expression, 10% (N=5) had isolated CD8 expression, dual expression for both CD4 and CD8 was observed in 45% (N=22) patients, and 20% (N=9) of the patients did not express either antigen. Expression frequency for myeloid/stem cell antigens (ETP-ALL vs. near-ETP-ALL patients) was CD117 (47% vs. 7%, P=0.011), CD34 (82% vs. 80%, P=0.865), HLA-DR (53% vs. 21%, P=0.073), CD13 (53% vs. 20%, P=0.055), CD33 (47% vs. 73%, P=0.131), and CD11b (29% vs. 21%, P=0.631).

Differences in the percentage of patients expressing immaturity associated antigens (CD10, CD34, and CD117), B-lineage antigens (CD19 and CD79a), myeloid antigens (CD13, CD11b, CD33), and non-lineage-specific antigens (CD123, CD56, and CD38) among our immunophenotypic subcategories of T-ALL are depicted in Fig. 1.

Figure 1. Percentage of patients expressing lineage-specific and non-specific antigens across the immunophenotypic subtypes of T-ALL.

EOI-MRD assessment

Among 60 patients who completed induction, EOI-MRD was tested in 59 (40 con-T-ALL, 12 ETP-ALL, and 7 near-ETP-ALL). A median of 2.3 million events (range, 0.18 to 7.3 million) was acquired for analysis, and over 1.5 million events were acquired in 68% of the samples.

EOI-MRD was positive in 39% of the samples tested (32% of pediatric and 52% of adult samples). EOI-MRD was frequently positive among ETP-ALL (75%, P=0.001) and near-ETP-ALL (71%, P=0.009) patients, compared to con- T-ALL patients (22.5%).

Median (range) MRD quantified among con-T-ALL, ETP-ALL, and near-ETP-ALL samples was 0.192% (0.015–2.125), 5.360% (0.125–30.306), and 4.250% (0.532–10.436), respectively. There was a significant difference in EOI-MRD quantified between con-T-ALL vs. near-ETP-ALL patients (P=0.019), but not between con-T-ALL vs. ETP-ALL (P=0.074) and ETP-ALL vs. near-ETP-ALL (P=0.898) patients. Comparison of the clinico-laboratory profiles of our EOI-MRD positive and negative T-ALL patients, and their 2-year OS, RFS, and EFS rates are presented in Supplementary Table 3.

Age group-specific analysis revealed significantly different frequencies of EOI-MRD positivity between the subcategories of T-ALL (con-T-ALL vs. ETP-ALL vs. near-ETP- ALL) among pediatric (15% vs. 83.3% vs. 60%, P=0.002), but not adult T-ALL patients (38.5% vs. 67% vs. 100%, P=0.190).

Antigen stability

Analysis of the effect of induction therapy on expressions of CD4, CD8, CD7, CD5, CD38, and sCD3 antigens revealed statistically significant upregulations of CD8 (P=0.046) and CD38 (P=0.046) expression in EOI blasts of con-T-ALL and ETP-ALL patients, respectively (Supplementary Fig. 3).

OS, RFS, and EFS

Among the 81 T-ALL patients, 4 died before treatment and 11 left hospital care before initiating treatment (refer to Supplementary Fig. 4 for disease course during follow-up). Among the remaining 66 patients who were treated, 6 died during the induction phase (4.7% of con-T-ALL, 7.6% of ETP-ALL, and 27% of near-ETP-ALL; P=0.068). Induction failure was observed in 8.3% of ETP-ALL, 30% of near- ETP-ALL, and none of our con-T-ALL patients (P=0.005). A mean (±SD) follow-up of 12 (±10) months was available among 60 patients who had completed induction. None of the patients underwent hematopoietic stem cell transplantation.

Irrespective of the age at diagnosis and immunophenotypic subclassification, 2-year OS, RFS, and EFS rates among our T-ALL patients were 65%, 76%, and 64.5%, respectively. The survival profiles of our T-ALL patients pertinent to their immunophenotypic subcategorization are depicted in Table 1 and Fig. 2. Expression of CD56, CD19, and CD79a in the blasts did not have any significant impact (P>0.05) on the OS, RFS, and EFS among any of the immuno-phenotypic subcategories of T-ALL. The 2-year OS, RFS, and EFS specific to our pediatric and adult patients stratified by T-ALL subtypes are summarized in Table 2 and compared in Supplementary Table 2.

Figure 2. Two-year overall survival (OS), relapse-free survival (RFS), and event-free survival (EFS) across all immunophenotypic subcategories of T-ALL analyzed together among all age groups (first row), pediatric patients (second row), and adult patients (third row).

Impact of EOI-MRD status on survival

Irrespective of immunophenotypic subclassification and age, there were significant differences in 2-year OS (86% vs. 48%, P=0.013), RFS (87% vs. 57%, P=0.022), and EFS (83% vs. 58%, P=0.008) between our EOI-MRD negative vs. positive T-ALL patients (refer to Supplementary Table 3 and Supplementary Fig. 5A for Kaplan–Meier survival curves).

Age group specific analysis revealed significant differences in 2-year OS (95% vs. 53%, P=0.044), RFS (95% vs. 60%, P=0.010), and EFS (95% vs. 60%, P=0.010) among EOI-MRD negative vs. positive pediatric T-ALL patients. However, 2-year OS (58% vs. 41%, P=0.303), RFS (67% vs. 54%, P=0.727) and EFS (50% vs. 44%, P=0.435) was not significantly different between EOI-MRD negative vs. positive adult T-ALL patients (refer to Supplementary Fig. 5B for Kaplan–Meier survival curves).

T-ALL subtype specific analysis revealed significant difference in 2-year OS (94% vs. 37%, P=0.012), RFS (94% vs. 50%, P=0.005), and EFS (94% vs. 50%, P=0.005) between EOI-MRD negative vs. positive pediatric con-T-ALL patients. However, there were no significant differences in 2-year OS (50% vs. 53%, P=0.874), RFS (62% vs. 66%, P=0.584) and EFS (42% vs. 53%, P=0.891) between the EOI-MRD negative vs. positive adult con-T-ALL patients (refer to Supplementary Fig. 5B for Kaplan–Meier survival curves). The lower number of patients available at the EOI timepoint in ETP-ALL (6 pediatric and 6 adult) and near-ETP-ALL (5 pediatric and 2) subtypes precluded analysis of age group specific impact of EOI-MRD status on survival in these categories.

Cox proportional hazard regression analysis was performed to identify risks incurred by the immunophenotypic subtype of T-ALL, presence of mediastinal mass and hyperleukocytosis at diagnosis, and D8BNC and EOI-MRD positive status on 2-year OS, RFS, and EFS on our pediatric and adult patients. The results are presented in Table 3.

Table 3 . Univariate analysis of covariates with event-free, relapse-free, and overall survivals..

Variables2 years-EFS2 years-RFS2 years-OS
HR95% CIPHR95% CIPHR95% CIP
Pediatric univariateD8BNC0.6560.119–3.6200.6290.8160.135–4.9240.8250.3110.035–2.8010.298
Mediastinal mass1.8490.358–9.5490.4633.1090.347–27.8430.3111.5780.285–8.7210.601
Hyper-leucocytosis0.4220.082–2.1790.3030.6930.116–4.1540.6880.2280.027–1.9510.117
EOI-MRD positive10.1531.132–91.0960.03810.0811.123–90.4950.0397.3810.757–71.9520.085
Con T-ALL subtype1.1290.219–5.8210.8850.7010.117–4.1970.6982.2720.265–19.4900.454
ETP-ALL subtype0.8870.103–7.1320.8871.2270.137–10.9840.8550.9320.108–8.0140.949
Near-ETP-ALL subtype0.9560.115–7.9470.9671.4250.159–12.7570.7620.7800.091–6.7000.821
Adult univariateD8BNC1.1660.326–4.1720.8140.4480.046–4.3360.4881.4560.388–5.4620.577
Mediastinal mass3.0000.782–11.5020.1092.2100.426–11.4620.3115.0081.029–24.3740.056
Hyper-leucocytosis1.7840.615–5.1780.2874.0840.908–18.3680.0671.4820.483–4.5470.491
EOI-MRD positive1.6480.461–5.8830.4421.3020.291–5.8280.7302.0240.501–8.1850.323
Con-T-ALL subtype0.4250.144–1.2530.1210.6070.135–2.7380.5160.3610.117–1.1170.077
ETP-ALL subtype0.7300.203–2.6230.6300.4610.055–3.8330.4730.8240.226–3.0020.769
Near-ETP-ALL subtype7.9952.000–31.9680.00311.1221.533–80.7190.0176.6491.891–23.3830.003

Abbreviations: CI, confidence interval; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; HR, hazard ratio; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival..


DISCUSSION

Demography

The 15% frequency of ETP-ALL documented in our pediatric T-ALL patients is similar to previously observed frequencies of 11% and 14% [4, 6]. As reported in other studies [17, 18], we too observed adult age predilection for ETP-ALL (P=0.039) in our cohort. ETP-ALL compromised 29% of our adult T-ALL patients. The documented frequency is variable, with rates of 17% in the United States, 32% in Germany, and 47% in China [1, 18-22]. This marked heterogeneity might be due either to ethnic predisposition or incongruencies in FCI analysis, where both ETP-ALL and near-ETP-ALL are considered under a common ETP-ALL category.

The exact worldwide frequency of near-ETP-ALL is unknown, as only a few studies have recognized this entity [3, 4, 6, 19]. In the present study, near-ETP-ALL was also frequent among adult T-ALL patients (26%, P=0.040). This frequency is similar to the 33% frequency observed by Van Vlierberghe et al. [19]. The 13% frequency of near-ETP-ALL we observed is similar to the 17% frequency reported by the Children’s Oncology Group [4], but is higher than the 5.4% frequency reported by Tembhare et al. [6] from India.

Our results indicate that these immunophenotypic subcategories of T-ALL cannot be distinguished by the presence of hepatosplenomegaly, lymphadenopathy, or mediastinal mass at diagnosis (P>0.05). Regarding the laboratory parameters at diagnosis, our adult ETP-ALL patients had significantly lower white blood cell (P=0.036) and higher platelet (P=0.016) counts compared to those in our adult con- T-ALL patients. These findings are consistent with those of prior studies [21, 22]. However, Ma et al. [23] had observed significantly low white blood cell counts among their pediatric ETP-ALL patients.

Immunophenotype at diagnosis

Both ETP-ALL and near-ETP-ALL blasts are proposed to have originated from BM-derived early thymic precursor (ETP) cells that migrated to the thymus. These ETP cells are too immature and have a transcriptome profile enabling differentiation towards T, myeloid, and dendritic cell lineages [1]. The dendritic-lineage orientation of ETP-ALL and near-ETP-ALL blasts was reflected in our results, as we observed a high frequency of CD123 positivity (P<0.001) in these patients as compared to con-T-ALL patients (Fig. 1). In our T-ALL cohort, CD73 expression was restricted only to ETP-ALL and near-ETP-ALL blasts (P=0.007). CD86 was significantly expressed only among ETP-ALL blasts (P< 0.001) (Fig. 1). The diagnostic relevance of this observation has to be verified in a larger cohort.

Regarding cross-lineage antigen expression among T-ALL blasts, expression of the CD56 antigen of natural killer (NK) cells is frequently associated with ETP-ALL blasts and confers a poor prognosis [24-27]. Consistent with the literature, we also observed a higher frequency of CD56 expression in our ETP-ALL and near-ETP-ALL patients (P=0.009). However, in contrast to the available literature, expression of CD56 did not make any difference in the OS, EFS, and RFS of any immunophenotypic subcategories of our T-ALL patients (P>0.05). This contrast might reflect the smaller cohort size, differences in treatment protocols used, and limited follow-up available among our patients. Despite recent observations regarding the expression of B-lineage markers CD19 and CD79a in ETP-derived blasts [6, 28], the prognostic relevance of such aberrant expression is unknown. In the current study, CD19 expression was observed only among ETP-ALL and near-ETP-ALL blasts and not among con- T-ALL blasts (P=0.004). In contrast, CD79a expression was not predilected towards any of the immunophenotypic subcategories of T-ALL (P=0.172). Importantly, aberrant expression of either CD19 or CD79a did not translate into inferior 2-year survival outcomes in our con-T-ALL, ETP-ALL, and near-ETP-ALL patient categories (P>0.05).

EOI-MRD

Traditional T-ALL MRD assessment by FCM relies on identifying the expression of immaturity associated markers like CD34, TdT, and CD99 on CD7 and cytoplasmic CD3 expressing lymphocytes [4, 8, 9, 29]. This approach is not foolproof as these immaturity-related antigens are frequently down-regulated during treatment [29]. T-ALL MRD analysis by FCM is also hindered by the presence of NK cells and their precursors that can mimic residual disease [6]. Due to these shortcomings, most T-ALL MRD data are by high-throughput sequencing for IgG and TCR rearrangements. The literature on T-MRD by FCM is limited [4, 8, 9, 29, 30]. However, these molecular MRD detection techniques might not be successful in ETP-ALL and near-ETP-ALL samples as these leukemias originate from precursor cells that are too immature to have undergone TCR rearrangement [3].

With the increased availability of ≥8 color flow cytometers, the results and sensitivity of T-MRD assessment by FCM are highly comparable to molecular T-MRD assays [8]. Use of 8–9 color panels by the Children’s Oncology Group (COG) yielded EOI-MRD detection rates of 30.5%, 81.4%, and 64.8% in pediatric (N=1,144) con-T-ALL, ETP-ALL, and near-ETP-ALL patients, respectively [4]. In an Indian study in which 35 T-ALL patients of all age groups were analyzed using an 8-color panel, EOI-MRD was detectable in 37% of the patients [30]. A recent study discussed the experience with T-ALL MRD using an 11-antigen 10-color FCM panel. Use of a mature T-cell antigen-based “exclusion” approach for gating detected EOI-MRD in 46.5% of the pediatric T-ALL cohort (N=269) [6]. In all these studies, cumulative MRD positivity observed among ETP-ALL and near-ETP-ALL patients was higher than the MRD positivity observed among con-T-ALL patients (74% with P<0.001 [6], 73.1% by the COG [4], and 67% with P=0.033 [30]). Our result was similar (73% with P<0.001).

Tembhare et al. [6] observed subtle down-regulation of surface CD3, CD4, CD5, CD8, and CD38 expression in their EOI-residual blasts. In the current study, we observed stable expression of CD4, CD5, CD7, and surface CD3 between the baseline and EOI-residual leukemic blasts (P>0.05) among all immunophenotypic subcategories of T-ALL. However, there was upregulated expression of CD8 (P=0.046) and CD38 (P=0.046) in the EOI blasts of con-T-ALL and ETP-ALL samples, respectively (Supplementary Fig. 3). Tembhare et al. [6] described that these immunophenotype shifts were too subtle to interfere with MRD analysis. Our findings are similar. These collective results indicate that the mature T-cell-related antigen-based approach is reliable for T-MRD analysis by FCM. Importantly, the expression of CD38 in nearly 98% of T-ALL patients at diagnosis in the present and previous [6] studies, along with the stable/upregulated expression of this antigen in the EOI-residual blasts, can be used as a potential target for daratumumab therapy [31]. In our cohort, CD73 expression was specifically associated with ETP-ALL and near-ETP-ALL patients (Fig. 2). Recent optimistic results were obtained with the use of anti-CD73 monoclonal antibody-based treatment in solid tumors [32]. Validation of the stability and specific expression of CD73 in ETP-ALL and near-ETP-ALL patients in a larger cohort would provide a rationale for initiating anti- CD73-based clinical trials in the treatment of these patients.

EOI-MRD status among our ETP-ALL and near-ETP-ALL patients was independent of their baseline clinical and laboratory characteristics. However, our EOI-MRD negative con-T-ALL patients were frequently diagnosed with mediastinal mass, high white blood cell count, and hyperleucocytosis at diagnosis as compared to those in the EOI-MRD positive counterparts.

Outcome in pediatric patients

Conter et al. [33] observed D8BNC status among 55% of their pediatric ETP-ALL patients. This result is similar to that in our cohort, where 50% of patients in each of the ETP-ALL and near-ETP-ALL categories were steroid unresponsive on day 8 (Table 2). High incidence of induction failure and disease relapse has been documented among pediatric ETP-ALL patients [2, 4, 23, 34]. However, none of our pediatric con-T-ALL, ETP-ALL, or near-ETP-ALL patients experienced induction failure or had a significantly higher relapse (Table 2). These contrasting observation have to be validated in a larger cohort with extended follow-up.

We observed no differences (P>0.05) in the 2-year OS, RFS, and EFS among our pediatric con-T-ALL (79%, 83%, and 81%, respectively), ETP-ALL (67%, 87%, and 80%), and near-ETP-ALL (100%, 75%, and 75%) patients. Consistently, there were no significant differences in 5-year OS and EFS among the con-T (92% and 86.9%, respectively), ETP-ALL (93% and 87%), and near-ETP-ALL (91.6% and 84.4%) patients documented in the largest ever pediatric T-ALL data by the Children’s Oncology Group [4]. However, inferior 2-, 4-, 5-, and 10-year OS and EFS have also been documented among pediatric-ETP-ALL patients from Italy, Japan, China, and USA, respectively [2, 23, 34]. These different observations might be due to heterogeneity in the sample sizes and ethnicities of the study cohorts. Hence, the true prognostic relevance of both ETP-ALL and near-ETP-ALL will be determined only by prospective studies with a large sample size and longer follow-up.

Irrespective of immunophenotypic sub classification, our EOI-MRD negative pediatric T-ALL patients had significantly better 2-year OS (95% vs. 53%, P=0.044), RFS (95% vs. 60%, P=0.010), and EFS (95% vs. 72%, P=0.010) than their EOI-MRD positive counterparts, consistent with previous observations [7].

Outcome in adult patients

In the current study, we did not observe any differences in 2-year OS, EFS, and RFS between our adult con-T-ALL and ETP-ALL patients (Table 2). These findings are consistent with two prior studies [21, 22], but discordant with the inferior OS documented among adult & adolescent ETP-ALL patients by Jain et al. [20]. Importantly, our adult near- ETP-ALL patients had the most inferior 2-year OS, EFS, and RFS as compared to that in the con-T-ALL and ETP-ALL patients diagnosed in this age group (Table 2).

An important observation from our study is the EOI-MRD positive status (irrespective of T-ALL subtype) and the presence of near-ETP-ALL immunophenotype (irrespective of EOI-MRD status) influencing the 2-year survival profile among our pediatric and adult T-ALL patients, respectively. These results are encouraging concerning hematopoietic stem cell transplantation at first remission in EOI-MRD positive pediatric T-ALL patients and all adult T-ALL patients diagnosed with near-ETP-ALL. However, the small age-specific sample size in our study precludes any definite conclusions.

CONCLUSION

Both ETP-ALL and near-ETP-ALL are common among adult T-ALL patients. Irrespective of age at diagnosis, both these entities are associated with a high frequency of EOI-MRD positivity. Our results indicate adverse 2-year survival conferred by the presence of EOI-MRD positivity among pediatric T-ALL patients and by the diagnosis of near-ETP-ALL phenotype among adult T-ALL patients. However, large prospective clinical trials are warranted to confirm these conclusions.

Study limitations

Being a relatively rare disease, the number of patients in our cohort might not be powered for exact outcome analysis across each immunophenotypic subcategory of T-ALL. Hence, the survival outcomes documented in our study have to be validated in a larger cohort. The mutational profile of leukemic lymphoblasts was not evaluated in our patients. Being retrospective data, our study results are purely observational and did not explore the underlying leukemogenic differences between the subtypes of T-ALL.

ACKNOWLEDGMENTS

Mrs. Arcot Radhakrishnan Abitha (Senior scientific assistant) is acknowledged for her efforts in FCM sample processing and sample acquisition. Dr. Rama R, assistant professor and statistician, helped with the statistics involved in the manuscript.

Authors’ Disclosures of Potential Conflicts of Interest

No potential conflicts of interest relevant to this article were reported.

Fig 1.

Figure 1.Percentage of patients expressing lineage-specific and non-specific antigens across the immunophenotypic subtypes of T-ALL.
Blood Research 2022; 57: 175-196https://doi.org/10.5045/br.2022.2022104

Fig 2.

Figure 2.Two-year overall survival (OS), relapse-free survival (RFS), and event-free survival (EFS) across all immunophenotypic subcategories of T-ALL analyzed together among all age groups (first row), pediatric patients (second row), and adult patients (third row).
Blood Research 2022; 57: 175-196https://doi.org/10.5045/br.2022.2022104

Table 1 . Clinical and laboratory characteristics of T-ALL subcategories..

ParametersOverall T-ALL (N=81)T-ALL subcategoriesP
Con-T-ALL
(N=49)
ETP-ALL
(N=17)
Near-ETP-ALL
(N=15)
ETP-ALL
vs.
Near-ETP-ALL
ETP-ALL
vs.
Con-T-ALL
Near-ETP-ALL
vs.
Con-T-ALL
Median (range) age in years17 (1–52)15 (1–50)17 (13–39)23 (5–52)0.8820.0030.016
Age group1.0000.0390.040
Pediatric (%)47 (58)34 (72%)7 (15%)6 (13%)
Adult (%)34 (42)15 (44%)10 (29%)9 (27%)
Sex (male:female)3.8:14.4:13.2:12.7:11.0000.6450.485
Median (range) Hb in g/L90 (30–142)90 (30–142)92 (30–131)88 (41–133)0.7371.0000.751
Median (range) WBC count, ×109/L64.1 (1–850)173 (1.1–850)70 (1–480)145 (3–590)0.0490.0050.751
Median (range) platelet, ×109/L54 (20–380)73 (20–366)125 (30–290)127 (20–380)0.9110.0080.080
Median (range) BM blast, %87 (22–99)87 (23–97)86 (22–98)89 (50–99)0.4730.7950.663
Median (range) PB blast, %78 (2–99)80 (2–97)42 (2–98)83 (2–99)0.1930.1740.411
Hyperleukocytosis41%45%18%53%0.0340.0460.567
Hepatomegaly42%42%27%58%0.2040.2830.319
Splenomegaly56%56%47%67%0.5160.5500.489
Lymphadenopathy78%73%87%86%1.0000.2900.342
Mediastinal mass31%36%33%13%0.3880.8420.095
CNS involvement at diagnosis3.2%2 (5)0%0%-0.4170.499
D8BNC35%32%54%20%0.2230.1680.440
EOI-MRD positive39% (N=59)22.5% (N=40)75%(N=12)71.4% (N=7)0.8650.0010.009
Relapse20% (N=60)18% (N=40)17% (N=12)38% (N=8)0.2920.9470.204
OS at 24 months65.2% (N=66)70.6% (N=42)60.4% (N=13)52% (N=11)0.1800.5510.019
RFS at 24 months76.1% (N=60)80% (N=40)79% (N=12)54.7% (N=8)0.2920.9560.190
EFS at 24 months64.5% (N=66)70.3% (N=42)66.6% (N=13)41% (N=11)0.0760.9780.013

Abbreviations: BM, bone marrow; CNS, central nervous system; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; Hb, hemoglobin; N, number of patients analyzed; NA, not applicable; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival; WBC, white blood cells..


Table 2 . Clinical and laboratory characteristics of immunophenotypic T-ALL subcategories among pediatric and adult age groups..

ParametersPediatric patientsAdult patients
T-ALL subtypePT-ALL subtypeP
Con-T-ALL
(N=34)
ETP-ALL
(N=7)
Near-ETP-ALL
(N=6)
ETP-ALL
vs.
Near- ETP-ALL
ETP-ALL
vs.
Con-T-ALL
Near- ETP-ALL
vs.
Con-T- T-ALL
Con-T-ALL
(N=15)
ETP-ALL
(N=10)
Near-ETP-ALL
(N=9)
ETP-ALL
vs.
Near- ETP-ALL
ETP-ALL
vs.
Con- T-ALL
Near- ETP-ALL
vs.
Con-T-ALL
Median age (range) in years12 (1–18)16 (13–17)13
(5–18)
0.2950.0560.34325
(20–50)
34
(19–39)
29
(20–52)
0.8420.3670.290
Sex (male:female)3.8:16:15:10.9060.3070.8256.5:12.3:12:10.8760.7020.243
Median (range) Hb in g/L91 (30–141)97
(30–131)
83
(41–129)
0.1810.4650.51789
(63–142)
80
(61–128)
88
(69–133)
0.3560.3381.000
Median (range) WBC, ×109/L110 (1.9–850)90.4
(3.2–267)
244
(3–590)
0.3660.1980.81088
(1.1–349)
55
(1–480)
68
(3.6–131)
0.0170.0360.682
Median (range) platelet, ×109/L83 (22–366)125
(30–245)
149
(32–380)
0.7310.1750.24052
(20–119)
125
(30–290)
100
(20–218)
0.7200.0160.138
Median (range) BM blast, %87 (23–97)86
(22–98)
95
(89–99)
0.1490.8450.02987
(64–96)
85
(38–95)
76
(50–97)
0.8630.7700.446
Median (range) PB blast, %84 (2–96)86
(2–98)
98
(2–99)
0.2680.8830.07461
(3–97)
36
(5–94)
76
(5–91)
0.1610.0730.770
Hyperleukocytosis53%29%67%0.0890.2400.53327%10%44%0.0890.3070.371
Hepatomegaly45%40%67%0.1090.8290.47836%20%56%0.1090.4040.349
Splenomegaly61.3%40%67&0.4630.3700.85543%50%67%0.4630.7290.265
Lymphadenopathy75%100%100%0.9060.2070.20770%80%78%0.9060.5600.658
Mediastinal mass40%20%17%0.1530.4020.28629%40%11%0.1530.5580.322
CNS involvement4%0%0%NA0.6380.6388%0%0%NA0.4520.620
Induction death7%0%0%0.0980.5080.5080%14%60%0.0980.1620.002
Induction failure0%0%0%NANANA0%17%75%0.0650.0010.001
D8BNC30%50%50%0.1050.3430.42240%57%0%0.1050.4860.074
EOI-MRD positive15%
(N=27)
83%
(N=6)
60%
(N=5)
0.3460.0010.02538.5%
(N=13)
67%
(N=6)
100%
(N=2)
0.3460.2520.104
Relapse11%
(N=27)
17%
(N=6)
17%
(N=6)
1.0000.7060.70631%
(N=13)
17%
(N=6)
100%
(N=2)
0.0350.2780.278
OS at 24 months79%
(N=29)
67%
(N=6)
100%
(N=6)
0.5640.8200.29748%
(N=13)
51%
(N=7)
0%
(N=5)
0.0250.5880.001
RFS at 24 months87%
(N=27)
83%
(N=6)
75%
(N=6)
0.9370.8050.72064%
(N=13)
75%
(N=6)
0%
(N=2)
0.0120.7050.014
EFS at 24 months81%
(N=29)
80%
(N=6)
75%
(N=6)
0.9370.8780.94345%
(N=13)
54%
(N=7)
0%
(N=5)
0.0190.767<0.001

Abbreviations: BM, bone marrow; CNS, the central nervous system; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; Hb, hemoglobin; N, number of patients analyzed; NA, not applicable; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival; WBC, white blood cells..


Table 3 . Univariate analysis of covariates with event-free, relapse-free, and overall survivals..

Variables2 years-EFS2 years-RFS2 years-OS
HR95% CIPHR95% CIPHR95% CIP
Pediatric univariateD8BNC0.6560.119–3.6200.6290.8160.135–4.9240.8250.3110.035–2.8010.298
Mediastinal mass1.8490.358–9.5490.4633.1090.347–27.8430.3111.5780.285–8.7210.601
Hyper-leucocytosis0.4220.082–2.1790.3030.6930.116–4.1540.6880.2280.027–1.9510.117
EOI-MRD positive10.1531.132–91.0960.03810.0811.123–90.4950.0397.3810.757–71.9520.085
Con T-ALL subtype1.1290.219–5.8210.8850.7010.117–4.1970.6982.2720.265–19.4900.454
ETP-ALL subtype0.8870.103–7.1320.8871.2270.137–10.9840.8550.9320.108–8.0140.949
Near-ETP-ALL subtype0.9560.115–7.9470.9671.4250.159–12.7570.7620.7800.091–6.7000.821
Adult univariateD8BNC1.1660.326–4.1720.8140.4480.046–4.3360.4881.4560.388–5.4620.577
Mediastinal mass3.0000.782–11.5020.1092.2100.426–11.4620.3115.0081.029–24.3740.056
Hyper-leucocytosis1.7840.615–5.1780.2874.0840.908–18.3680.0671.4820.483–4.5470.491
EOI-MRD positive1.6480.461–5.8830.4421.3020.291–5.8280.7302.0240.501–8.1850.323
Con-T-ALL subtype0.4250.144–1.2530.1210.6070.135–2.7380.5160.3610.117–1.1170.077
ETP-ALL subtype0.7300.203–2.6230.6300.4610.055–3.8330.4730.8240.226–3.0020.769
Near-ETP-ALL subtype7.9952.000–31.9680.00311.1221.533–80.7190.0176.6491.891–23.3830.003

Abbreviations: CI, confidence interval; D8BNC, day 8 blast not cleared; EFS, event-free survival; EOI-MRD, end-of-induction-measurable residual disease; HR, hazard ratio; OS, overall survival; PB, peripheral blood; RFS, relapse-free survival..


References

  1. Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. Revised 4th ed. Lyon, France:. IARC Press,:421.
  2. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. Lancet Oncol 2009;10:147-56.
    Pubmed KoreaMed CrossRef
  3. Wu D, Sherwood A, Fromm JR, et al. High-throughput sequencing detects minimal residual disease in acute T lymphoblastic leukemia. Sci Transl Med 2012;4:134ra63.
    Pubmed CrossRef
  4. Wood BL, Winter SS, Dunsmore KP, et al. T-lymphoblastic leukemia (T-ALL) shows excellent outcome, lack of significance of the early thymic precursor (ETP) immunophenotype, and validation of the prognostic value of end-induction minimal residual disease (MRD) in Children's Oncology Group (COG) Study AALL0434. Blood (ASH Annual Meeting Abstracts) 2014;124:1.
    CrossRef
  5. Lin N, Liu ZH, Wang PP, Fu W, Yan XJ, Li Y. Immunophenotypic analysis of patients with adult acute T-lymphoblastic leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2020;28:442-5.
    Pubmed CrossRef
  6. Tembhare PR, Chatterjee G, Khanka T, et al. Eleven-marker 10-color flow cytometric assessment of measurable residual disease for T-cell acute lymphoblastic leukemia using an approach of exclusion. Cytometry B Clin Cytom 2021;100:421-33.
    Pubmed CrossRef
  7. Tembhare PR, Narula G, Khanka T, et al. Post-induction measurable residual disease using multicolor flow cytometry is strongly predictive of inferior clinical outcome in the real-life management of childhood T-cell acute lymphoblastic leukemia: a study of 256 patients. Front Oncol 2020;10:577.
    Pubmed KoreaMed CrossRef
  8. Modvig S, Madsen HO, Siitonen SM, et al. Minimal residual disease quantification by flow cytometry provides reliable risk stratification in T-cell acute lymphoblastic leukemia. Leukemia 2019;33:1324-36.
    Pubmed CrossRef
  9. Thörn I, Forestier E, Botling J, et al. Minimal residual disease assessment in childhood acute lymphoblastic leukaemia: a Swedish multi‐centre study comparing real‐time polymerase chain reaction and multicolour flow cytometry. Br J Haematol 2011;152:743-53.
    Pubmed CrossRef
  10. Gudapati P, Khanka T, Chatterjee G, et al. CD304/neuropilin‐1 is a very useful and dependable marker for the measurable residual disease assessment of B‐cell precursor acute lymphoblastic leukemia. Cytometry B Clin Cytom 2020;98:328-35.
    Pubmed CrossRef
  11. Manabe A, Ohara A, Hasegawa D, et al. Significance of the complete clearance of peripheral blasts after 7 days of prednisolone treatment in children with acute lymphoblastic leukemia: the Tokyo Children's Cancer Study Group Study L99-15. Haematologica 2008;93:1155-60.
    Pubmed CrossRef
  12. Cherian T, John R, Joseph LL, et al. Complete peripheral blast clearance is superior to the conventional cut-off of 1000/µL in predicting relapse in pediatric pre-B acute lymphoblastic leukemia. Indian J Hematol Blood Transfus 2021;37:366-71.
    Pubmed KoreaMed CrossRef
  13. Bommannan BKK, Arumugam JR, Sundersingh S, Rajan PT, Radhakrishnan V, Sagar TG. CD19 negative and dim precursor B-lineage acute lymphoblastic leukemias: real-world challenges in a targeted-immunotherapy era. Leuk Lymphoma 2019;60:3154-60.
    Pubmed CrossRef
  14. Dworzak MN, Buldini B, Gaipa G, et al. AIEOP‐BFM consensus guidelines 2016 for flow cytometric immunophenotyping of pediatric acute lymphoblastic leukemia. Cytometry B Clin Cytom 2018;94:82-93.
    Pubmed CrossRef
  15. Chopra A, Bakhshi S, Pramanik SK, et al. Immunophenotypic analysis of T‐acute lymphoblastic leukemia. A CD 5‐based ETP‐ALL perspective of non‐ETP T‐ALL. Eur J Haematol 2014;92:211-8.
    Pubmed CrossRef
  16. Sędek Ł, Theunissen P, Sobral da Costa E, et al. Differential expression of CD73, CD86 and CD304 in normal vs. leukemic B-cell precursors and their utility as stable minimal residual disease markers in childhood B-cell precursor acute lymphoblastic leukemia. J Immunol Methods 2019;475:112429.
    Pubmed CrossRef
  17. Haydu JE, Ferrando AA. Early T-cell precursor acute lymphoblastic leukaemia. Curr Opin Hematol 2013;20:369-73.
    Pubmed KoreaMed CrossRef
  18. Neumann M, Heesch S, Gökbuget N, et al. Clinical and molecular characterization of early T-cell precursor leukemia: a high-risk subgroup in adult T-ALL with a high frequency of FLT3 mutations. Blood Cancer J 2012;2:e55.
    Pubmed KoreaMed CrossRef
  19. Van Vlierberghe P, Ambesi-Impiombato A, Perez-Garcia A, et al. ETV6 mutations in early immature human T cell leukemias. J Exp Med 2011;208:2571-9.
    Pubmed KoreaMed CrossRef
  20. Jain N, Lamb AV, O'Brien S, et al. Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype. Blood 2016;127:1863-9.
    Pubmed KoreaMed CrossRef
  21. Zhang Y, Qian JJ, Zhou YL, et al. Comparison of early T-cell precursor and non-ETP subtypes among 122 Chinese adults with acute lymphoblastic leukemia. Front Oncol 2020;10:1423.
    Pubmed KoreaMed CrossRef
  22. Liao HY, Sun ZY, Wang YX, Jin YM, Zhu HL, Jiang NG. Outcome of 126 adolescent and adult T-cell acute leukemia/lymphoma patients and the prognostic significance of early T-cell precursor leukemia subtype. Zhonghua Xue Ye Xue Za Zhi 2019;40:561-7.
    Pubmed KoreaMed CrossRef
  23. Ma M, Wang X, Tang J, et al. Early T-cell precursor leukemia: a subtype of high risk childhood acute lymphoblastic leukemia. Front Med 2012;6:416-20.
    Pubmed CrossRef
  24. Montero I, Rios E, Parody R, Perez-Hurtado JM, Martin-Noya A, Rodriguez JM. CD56 in T-cell acute lymphoblastic leukaemia: a malignant transformation of an early myeloid-lymphoid progenitor? Haematologica 2003;88:ELT26.
    Pubmed
  25. Fischer L, Gökbuget N, Schwartz S, et al. CD56 expression in T-cell acute lymphoblastic leukemia is associated with non-thymic phenotype and resistance to induction therapy but no inferior survival after risk-adapted therapy. Haematologica 2009;94:224-9.
    Pubmed KoreaMed CrossRef
  26. Fuhrmann S, Schabath R, Möricke A, et al. Expression of CD56 defines a distinct subgroup in childhood T‐ALL with inferior outcome. Results of the ALL‐BFM 2000 trial. Br J Haematol 2018;183:96-103.
    Pubmed CrossRef
  27. Dalmazzo LF, Jácomo RH, Marinato AF, et al. The presence of CD56/CD16 in T‐cell acute lymphoblastic leukaemia correlates with the expression of cytotoxic molecules and is associated with worse response to treatment. Br J Haematol 2009;144:223-9.
    Pubmed CrossRef
  28. Garg S, Gupta SK, Bakhshi S, Mallick S, Kumar L. ETP-ALL with aberrant B marker expression: case series and a brief review of literature. Int J Lab Hematol 2019;41:e32-7.
    Pubmed CrossRef
  29. Roshal M, Fromm JR, Winter S, Dunsmore K, Wood BL. Immaturity associated antigens are lost during induction for T cell lymphoblastic leukemia: implications for minimal residual disease detection. Cytometry B Clin Cytom 2010;78:139-46.
    Pubmed KoreaMed CrossRef
  30. Singh N, Agrawal N, Sood R, et al. T-ALL minimal residual disease using a simplified gating strategy and its clinico-hematologic correlation: a single center experience from North India. Indian J Hematol Blood Transfus 2019;35:707-10.
    Pubmed KoreaMed CrossRef
  31. Tembhare PR, Sriram H, Khanka T, et al. Flow cytometric evaluation of CD38 expression levels in the newly diagnosed T-cell acute lymphoblastic leukemia and the effect of chemotherapy on its expression in measurable residual disease, refractory disease and relapsed disease: an implication for anti-CD38 immunotherapy. J Immunother Cancer 2020;8:e000630.
    Pubmed KoreaMed CrossRef
  32. Chen S, Wainwright DA, Wu JD, et al. CD73: an emerging checkpoint for cancer immunotherapy. Immunotherapy 2019;11:983-97.
    Pubmed KoreaMed CrossRef
  33. Conter V, Valsecchi MG, Buldini B, et al. Early T-cell precursor acute lymphoblastic leukaemia in children treated in AIEOP centres with AIEOP-BFM protocols: a retrospective analysis. Lancet Haematol 2016;3:e80-6.
    Pubmed CrossRef
  34. Inukai T, Kiyokawa N, Campana D, et al. Clinical significance of early T‐cell precursor acute lymphoblastic leukaemia: results of the Tokyo Children's Cancer Study Group Study L99‐15. Br J Haematol 2012;156:358-65.
    Pubmed CrossRef
Blood Res
Volume 59 2024

Supplementary File

Stats or Metrics

Share this article on

  • line

Related articles in BR

Blood Research

pISSN 2287-979X
eISSN 2288-0011
qr-code Download