scholarly journals Activated Natural Killer Cells Predict Poor Clinical Prognosis in High-risk B- and T- cell Acute Lymphoblastic Leukemia

Blood ◽  
2021 ◽  
Author(s):  
Caroline Duault ◽  
Anil Kumar ◽  
Adeleh Taghi Khani ◽  
Sung June Lee ◽  
Lu Yang ◽  
...  

B- and T- cell acute lymphoblastic leukemia (B/T-ALL) may be refractory or recur after therapy by suppressing host anti-cancer immune surveillance mediated specifically by natural killer (NK) cells. We delineated the phenotypic and functional defects in NK cells of high-risk B/T-ALL patients using mass, flow, and in silico cytometry, with the goal of further elucidating the role of NK cells in sustaining ALL regression. We found that, compared to normal counterparts, NK cells in B/T-ALL patients are less cytotoxic, but exhibit an activated signature characterized by high CD56, high CD69, production of activated NK-origin cytokines, and calcium signaling. We demonstrated that defective maturation of NK cells into cytotoxic effectors prevents NK cells of ALL patients from lysing NK-sensitive targets as efficiently as normal NK cells. Additionally, we showed that NK cells in ALL are exhausted, which is likely caused by their chronic activation. We found that increased frequencies of activated cytokine-producing NK cells are associated with increased disease severity and independently predict poor clinical outcome in ALL patients. Our studies highlight the benefits of developing NK cell profiling as a diagnostic tool to predict clinical outcome in patients with ALL and underscore the clinical potential of allogeneic NK infusions to prevent ALL recurrence.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 39-39
Author(s):  
Caroline Duault ◽  
Anil Kumar ◽  
Adeleh Taghi Khani ◽  
Sung June Lee ◽  
Lu Yang ◽  
...  

Background: High-risk B- and T- cell Acute Lymphoblastic Leukemia (B/T-ALL) are aggressive lymphoid malignancies being treated with inhibitors targeting oncogenes and CAR-T cell-based immunotherapies. Despite advances in treatment, emergence of therapeutic resistance poses a clinical challenge in combating B/T-ALL. More recently, NK cells have emerged as attractive immunotherapies for cancers. Furthermore, NK therapies show promise for T-ALL, that has been particularly hard to treat with CAR-T cells. Approach: To develop NK therapies against B/T-ALL, it is important to identify the phenotypic and functional deficiencies in NK cells of ALL patients. Therefore, using mass cytometry (CyTOF), we compared the profile of NK cells in bone marrow (BM) or peripheral blood (PB) of 20 high-risk ALL patients (13 B-ALL, 7 T-ALL, 9 pediatric and 11 adult) and 22 tissue-matched healthy donors. We validated our CyTOF results by conducting in silico cytometry (CIBERSORT) for NK subsets in an independent panel of 207 high-risk B-ALL patients from the Children's Oncology Group (COG) P9906 clinical trial. We then investigated whether distinct NK signatures identified by CyTOF and CIBERSORT in B/T-ALL correlate with risk features and predict clinical outcome. Results: We observed a significant reduction in the frequency of total NK cells within the non-malignant immune fraction in B/T-ALL. Upon comparing the frequencies of specific NK subsets (CD56Brightand CD56Dim, we found that CD56Bright NK cells were increased in the BM (P=0.0138) but were reduced in PB (P=0.0285) of patients as compared to healthy donors. We then conducted CIBERSORT to estimate the relative proportions of NK cells with CD56Bright and CD56Dim molecular signatures in 207 COG B-ALL patients compared to 94 healthy donors. We found that frequencies of NK cells with CD56Bright molecular signature were increased in both BM and PB of ALL patients (P<0.0001). We conclude that majority of NK cells in B/T-ALL exhibit the molecular profile of CD56Bright NK cells, albeit their PB NK cells show reduced surface CD56. Human NK cells mature from CD56Bright to CD56Dim stages. CD56Dim NK are more cytotoxic and produce less cytokines than CD56Bright NK cells. Enrichment of CD56Bright molecular signature in NK cells of B/T-ALL patients suggested an impairment in NK maturation and function in ALL. By comparing the abilities of PB NK cells in B/T-ALL and healthy donors to lyse K562 targets invitro, we found that NK cells in ALL patients are poorly cytolytic compared to healthy donors (P=0.0295). Upon stimulation with PMA/Ionomycin, we observed an increase in the frequencies of GM-CSF+ and TNF-α+ cells in total NK and NK subsets in BM and PB of B/T-ALL patients compared to healthy donors (all P<0.05). Surprisingly, the natural cytotoxicity receptor NKp46 was increased in stimulated NK subsets of B/T-ALL patients compared to healthy controls (P<0.01). We conclude that poorly cytotoxic NK cells with CytokineHigh NKp46High activated phenotype (NKActiv) are enriched in B/T-ALL, while normal NK phenotype (NKRest) is suppressed. Finally, we investigated whether relative frequencies of NKActiv and NKRest cells can predict clinical outcome in high-risk ALL. To this end, we used CIBERSORT to estimate proportions of NK cells with activated (NKActiv) and resting (NKRest) molecular signatures in 207 COG B-ALL patients. We then separated patients into 2 groups as NKActiv> NKRest and NKRest> NKActiv, and compared their relapse-free survival (RFS) probabilities (Fig.1). We found that patients with higher NKActiv cells had shorter RFS than those with higher NKRest cells (P=0.0028). Furthermore, we found that relative proportions of activated and resting NK cells independently predict clinical outcome within a poorly prognostic subset of patients with Central Nervous System involvement (CNS+, P=0.0098). These preliminary findings suggest that higher levels of activated NK cells in B-ALL may be associated with poor clinical prognosis. We are validating these results in B/T-ALL from our CyTOF studies. Conclusion: We find that impairment of NK maturation in high-risk human B/T-ALL results in the accumulation of NK cells with a poorly cytotoxic but hyperactivated cytokine-producing phenotype, that positively correlates with poor clinical prognosis. Our data suggest that restoring NK homeostasis would be an attractive strategy for treating high-risk B/T-ALL. Disclosures Marcucci: Novartis: Speakers Bureau; Abbvie: Speakers Bureau; Iaso Bio: Membership on an entity's Board of Directors or advisory committees; Takeda: Other: Research Support (Investigation Initiated Clinical Trial); Merck: Other: Research Support (Investigation Initiated Clinical Trial); Pfizer: Other: Research Support (Investigation Initiated Clinical Trial).


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 133-133 ◽  
Author(s):  
Barbara De Moerloose ◽  
Stefan Suciu ◽  
Alina Ferster ◽  
Françoise Mazingue ◽  
Nicolas Sirvent ◽  
...  

Abstract Abstract 133 Background: T-cell acute lymphoblastic leukemia (ALL) accounts for 15% of ALL cases in children and has been associated with a higher risk for central nervous system (CNS) relapse and a worse prognosis. In EORTC trials 58831 and 2, standard risk (SR) patients (pts) were not irradiated but received intermediate dose methotrexate (MTX) courses; for medium and high risk pts, high dose (HD) MTX was added to the treatment regimen and the administration of cranial radiotherapy (RT) was randomised. The omission of RT didn't result in an increase of CNS or systemic relapse and consequently, CNS-directed chemotherapy was substituted for RT in all following trials. The long-term outcome of T-ALL pts in the subsequent phase III trials (58881 and 58951) are presented here. Methods: The BFM backbone for ALL treatment was applied to all EORTC-CLG trials since 1983. As CNS treatment in study 58881, SR pts received 4 HD MTX courses (5 g/m2) in interval therapy and 10 IT MTX injections during the intensive treatment phases. Pts with CNS-3 status at diagnosis received 2 additional IT injections during induction, 2 during consolidation and 6 HD MTX courses + IT during maintenance. T-ALL pts with poor prephase response (PPR) at day 8 or who didn't achieve complete remission (CR) after induction were included in the very high risk (VHR) group. VHR CNS-directed chemotherapy included 10 IT MTX injections, 6 IT triple and 10 HD MTX courses during intensive treatment phases, followed by 4 IT MTX injections during maintenance (the latter for CNS-3 pts only). In the 58951 trial, all T-ALL pts had an intensified induction. The CNS-directed therapy of all average risk T-ALL pts was intensified to 11 HD MTX courses, 1 IT with MTX and 15 triple IT. MRD ≥ 1% at the end of induction was added as VHR criterium. All non-transplanted VHR pts received 1 IT MTX injection, 19 IT triple and 9 HD MTX courses. Several randomized questions were addressed in both trials of which most relevant for T-ALL pts: in study 58881 the comparison E.coli asparaginase (ASP) Medac versus (vs) “other ASP” (= Erwinia ASP or E.coli ASP Bayer); in trial 58951 1) the comparison dexamethasone (DEX 6 mg/m2/d) vs prednisolone (PRED 60 mg/m2/d) in induction and 2) conventional vs prolonged E.coli ASP for non-VHR pts. Results: 303 and 296 T-ALL pts were included in trials 58881 and 58951 resp, representing 14.5% and 15.2% of all pts. Outcome results and type of events for the entire 58881 and 58951 cohorts and according to several subgroups are presented in the table. The 8-year isolated and overall CNS relapse incidences were 6.8% and 10.9% in study 58881, 5.3% and 8.5% in study 58951. The 8-year EFS, DFS and OS improved remarkably in study 58951. In the latter trial, outcome improvement was particularly seen in pts with initial WBC<100x10E9/L and in the good prephase responders (GPR) which had a significant better outcome than those with PPR. 58881 pts assigned to the “other ASP” arm had an inferior outcome. Concerning the DEX/PRED comparison in the 58951 T-ALL cohort, no advantage was seen for EFS (hazard ratio (HR) (99%CI): 1.26 (0.70;2.27)) or OS. There even was a trend towards worse EFS for pts with initial WBC>100x10E9/L and for pts with PPR treated in the DEX arm (HR (99%CI): 1.52 (0.63;3.64) and 1.47 (0.64;3.35)). Prolonged ASP treatment did not improve outcome of the whole T-ALL 58951 cohort. Conclusion: Prophylactic and therapeutic RT can safely be omitted from frontline treatment of children with T-ALL. Adequate ASP therapy, intensified induction treatment and CNS directed therapy can result in a significant improvement of the outcome of at least 2/3rd of T-ALL pts, particularly those with initial WBC<100x10E9/L and GPR. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (1) ◽  
pp. 74-82 ◽  
Author(s):  
Pieter Van Vlierberghe ◽  
Alberto Ambesi-Impiombato ◽  
Kim De Keersmaecker ◽  
Michael Hadler ◽  
Elisabeth Paietta ◽  
...  

Key Points Integrated genomic profiling identifies high-risk adult T-ALL patients with poor response to intensified chemotherapy.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1083-1083 ◽  
Author(s):  
Arnaud Petit ◽  
Amélie Trinquand ◽  
Sylvie Chevret ◽  
Paola Fabiola Ballerini ◽  
Jean-Michel Cayuela ◽  
...  

Abstract Background: Risk stratification in childhood T-cell acute lymphoblastic leukemia (T-ALL) is crucial to drive treatment decisions. Since patients with induction failure or relapse are often refractory to further treatment, identifying high risk patients up-front will allow improved treatment. While minimal residual disease (MRD) is the strongest prognosis risk factor used after complete remission (CR), NOTCH1/FBXW7 (N/F) and RAS/PTEN (R/P) mutation profiles at diagnosis have recently been identified to predict outcome in adult T-ALL. Objective: to test whether an oncogenetic classifier using N/F and R/P mutations could improve the detection of children with T-ALL at risk of relapse. Methods: 405 patients with T-ALL aged from 1 to 14 years were treated according to FRALLE T guidelines (FRALLE Study group) between 2000 and 2010. Among them, 220 patients, for whom biological material at diagnosis was available, were tested retrospectively for N/F and R/P mutations. These study cohort patients were representative of overall FRALLE 2000 T-ALLs. CR was achieved in 213 patients. MRD (IgH-TCR markers) tested at CR (day 35) was available for 191 patients. MRD was <10-4 for 114 patients (60%) and ≥10-4 for 77 patients. Patients with N/F mutation and R/P germline (GL) were defined as oncogenetic low risk (LoR), while N/F GL and R/P GL or mutation and N/F mutation and R/P mutation were defined as high risk (HiR). Results: 111 patients were classified as LoR and 109 as HiR. Five-year-CIR and DFS were respectively 35.5% (95% CI, 26.7-44.3) and 59% (95%CI, 50.2-69.6) for HiR versus 13% (95% CI, 6.8-19.2) and 86.8% (80.5-93.5) for the LoR group (Figures A and B). HiR patients were significantly associated with MRD ≥ 10-4 (p=0.0004) and higher risk of relapse (p=0.00002). Among patients with MRD ≥ 10-4, HiR feature worsened the risk of relapse: 5-year-CIR and DFS were respectively 42.8% (95% CI, 28.9-56.7) and 71.1% (95%CI, 56.0-90.2) in HiR versus 28.9% (95% CI, 11.7-46.1) and 50.9% (95%CI, 38.4-67.6) in the LoR group. Among patients with MRD <10-4, 5-year-CIR and DFS were respectively 28.9 % (95% CI, 15.0-42.8) and 71.0% (95%CI, 58.4-86.3) in HiR group versus 4.4% (95% CI, 0-9.2) and 95.5% (95%CI, 90.7-1.00) in LoR group (Figures C and D). As such, the classifier allowed identification of 63% of very low risk patients amongst the MRD<10-4 population. Prognostic values of new oncogenetic risk factors were then analyzed with conventional factors. By univariate analysis, factors identified to predict relapse were male gender (p=0.036), WBC count ≥ 200 G/L (p=0.023), chemoresistance at day 21 (p=0.007), MRD ≥10-4 (p=0.0006) and oncogenetic HiR (p<0.0001). A multivariable cox model including these variables selected the classifier together with WBC count, day 21 chemo-sensitivity and MRD. Based on a stepwise selection procedure, the three most discriminating variables were classifier, WBC count and MRD. The cause specific Hazard Ratio (HR) was 3.22 (95% CI, 1.64-6.28) for oncogenetic HiR versus LoR (p=0.0006), 2.30 (95% CI, 1.26-4.20) for MRD≥10-4 versus MRD<10-4(p=0.0070) and 1.85 (95% CI, 1.01-3.37) for WBC≥200G/L versus <200 G/L (p=0.0456). Based on these three parameters, 8 subsets of patients were defined according to the estimated 5-year CIR. The 58 patients (30%) associating WBC count < 200G/L, classifier LoR and MRD<10-4 were at very low risk of relapse, with a 5-y-CIR of 1.7%. Patients harboring at least one of: WBC count ≥200G/L, classifier HiR or MRD>10-4, demonstrated an increasing CIR, up to 45.8% if all three were associated. Conclusion: in childhood T-ALL, oncogenetic classification using N/F and R/P mutation profiles is an independent predictor of relapse. When combined with MRD and WBC count ≥200 G/L, it significantly improved relapse prediction, particularly amongst the 60% of T-ALLs with MRD <10-4 at day 35. Appropriate integrating these 3 factors, will help optimize treatment. Figure Figure. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Tobias M. Dantonello ◽  
Mutlu Kartal-Kaess ◽  
Christoph Aebi ◽  
Franziska Suter-Riniker ◽  
Jasmin D. Busch ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4389-4389
Author(s):  
Johann Greil ◽  
Elke Kleideiter ◽  
Matthias Schwab ◽  
Petra Boukamp ◽  
Ewa Koscielniak ◽  
...  

Abstract Shortened telomeres and elevated levels of telomerase activity are apparently characteristic features of hematologic neoplasias such as high-grade lymphomas and relapsing leukemia. Thus, their measurement might be useful for monitoring disease conditions or predicting clinical outcome. In order to investigate the potential of telomere length (TRF) and telomerase activity (TA) as prognostic indicator in pediatric patients with T-cell acute lymphoblastic leukemia (T-ALL) we analyzed TRF and TA in samples from 20 patients (age range 2–17.5 years). In addition, as TA is limited by the expression of the telomerase catalytic subunit (hTERT) we analyzed hTERT expression. We found that TRF varied widely (3.5 – 8.1 kb; mean ± SD: 6.4 +/− 1.3 kb) in leukemic cells and was significantly shorter (p<0.0001) than that of age-matched controls (8.3 ± 0.4 kb; n=19). Elevated levels of TA were present in 95% of the leukemic samples. Furthermore, expression of hTERT demonstrated a wide interindividual variability (range 141–424,000 normalized units). A statistically significant association between TA and hTERT expression was not found and TRF, TA and hTERT expression was not associated with the clinical outcome in pediatric T-ALL, thereby limiting their prognostic significance.


Sign in / Sign up

Export Citation Format

Share Document