Absolute Lymphocyte Count Is a Novel Prognostic Indicator in Acute Leukemia: Implications for Risk Stratification and Future Studies.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 2276-2276
Author(s):  
Guillermo R. De Angulo ◽  
Carrie Yuen ◽  
Shana Palla ◽  
Peter M. Anderson ◽  
Patrick A. Zweidler-McKay

Abstract Background: Despite improving outcomes, 25–50% of children and young adults with acute leukemia still relapse and most salvage rates are discouraging. Additional prognostic factors, particularly those that represent host factors, may further stratify patients and decrease relapse rates. Purpose: To determine if absolute lymphocyte counts (ALC) during induction chemotherapy can improve current risk stratification and predict relapse-free survival (RFS) and overall survival (OS) in children and young adults with acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). Methods: We analyzed 160 consecutive cases of de novo ALL and AML patients 1–21 years of age, treated at the University of Texas M. D. Anderson Cancer Center from 1995–2005. Age at diagnosis, initial WBC, bone marrow blast % on days 0 and 7, were analyzed with ALC on days 0, 15, 21 and 28 of induction therapy. Results: ALC during induction therapy is a significant independent predictor of RFS and OS in young adults and children with either ALL or AML. Specifically, an ALC <350 cells/mcL on day 15 of induction therapy for ALL significantly predicts poor 6-year OS (52% vs. 87%, p=0.015; HR=4.2, Figure 1A) and RFS (46% vs. 80%, p=0.001; HR=4.8, Figure 1B). Similarly, an ALC of <350 cells/mcL on day 15 of induction therapy for AML predicts poor 6-year OS (35% vs. 86%, p=0.033; HR=4, Figure 1C). ALC-15 remains a significant predictor of OS and RFS after adjusting for age at diagnosis, initial WBC and bone marrow response on day 7 (p=0.013; HR=6.3, and p=0.003; HR=6.3, respectively) in multivariate analysis (Table 1). Importantly, ALC-15 defines a subgroup of half of our AML patients and predicts an excellent 5-year OS of 86% (p=0.033, Figure 1C). Conversely, prolonged lymphopenia predicts that 16% of young AML patients will have a dismal 5-year RFS of 14% (p=0.004, Figure 1D). Finally, ALC-15 <350 cells/mcL is able to predict 70% of relapses in both ALL and AML patients. One possible algorithm could identify half of AML patients with a predicted OS of 86% simply by measuring the ALC-15. Those patients with a low ALC on day 15 would be assessed at day 21 and 28 and those with persistent lymphopenia would be predicted to to have an RFS of 14% and would be stratified to receive intensified and/or experimental therapy. Conclusion: We demonstrate that ALC can identify patients at high and low risk for relapse early in the course of treatment for ALL or AML. Our data indicates that ALC is both independent of and a more powerful predictor than age at diagnosis, initial WBC and bone marrow response on day 7. This routine measurement could enhance current risk-stratification and lead to improved outcomes in young patients with acute leukemias. Figure 1 Figure 1. Table 1 Multivariate Analysis of ALC, Age, WBC, Bone marrow response and Survival

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2264-2264
Author(s):  
Daiichiro Hasegawa ◽  
Satoshi Hirase ◽  
Hironobu Takahashi ◽  
Atsuro Saito ◽  
Aiko Kozaki ◽  
...  

Abstract BACKGROUND: Recently, several studies have demonstrated that absolute lymphocyte counts (ALC) after induction therapy predicted treatment outcome. To address this issue, we here assessed the impact of the ALC at the end of induction therapy on outcomes in childhood ALL. METHODS: We reviewed 141 cases of pediatric ALL with 1-21 years of age treated on the Japan Association Childhood Leukemia Study group ALL-02 series of treatment trials between 2002 and 2013. Patients with Philadelphia chromosome-positive ALL were excluded. Variables retrospectively analyzed included ALC at several time points during remission induction, age at diagnosis, gender, initial white blood cell count (WBC), cytogenetics, immunological phenotype, stratified risk, treatment response for bone marrow (the percentage of blasts at day 15), and outcome. Events in the analysis of event-free survival (EFS) included induction failure, death, relapse and secondary malignant neoplasm. The comparison of categorical variables between groups was performed by chi-square test. The probability of EFS and overall survival (OS) were analyzed with the use of the Kaplan–Meier method and a stratified log-rank test. A multivariate analysis of survival was performed with the use of a Cox proportional-hazard model to evaluate the treatment effect with adjustment for stratification factors. RESULTS: The subjects included 121 of B-precursor ALL, 10 of T cell ALL, and 4 of acute mixed lineage leukemia/ acute unclassified leukemia. We found high WBC count at diagnosis (>100K/microL) and slow early responder for bone marrow at day 15 to be an unfavorable prognostic indicator, and also the ALC at the end of induction (day29) to be a statistically significant predictor of improved OS and EFS in our cohort. Patients with ALC ≥ 800/microL had a superior 5-year overall survival (100 ± 1.7% vs 88.1 ± 4.3 %, p=0.0001) and EFS (98.3 ± 1.7% vs 81.8 ± 5.0 %, p=0.0001). Multivariate analysis demonstrated that ALC at day29 was an independent, clinically significant predictor of improved EFS and OS after controlling WBC at diagnosis, gender, age at diagnosis, and cytogenetics. Multiple regression analysis adjusting for initial WBC count, peripheral blast counts at day8, and cytogenetics, also revealed an independent relationship (p=0.005) between treatment response (the percentage of blasts at day 15) and ALC at day29. CONCLUSIONS: ALC is a simple, statistically significant prognostic factor in childhood ALL that may refine current risk stratification algorithms. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3650-3650
Author(s):  
Natasha Szuber ◽  
Judith Jolin ◽  
Michael Harnois

Abstract Background: The median age at diagnosis of myeloproliferative neoplasms (MPN) is in the sixth decade, though adolescents and young adults (AYA) with MPN comprise a distinct, growing sub-population. Preliminary data has exposed a unique phenotype and clinical trajectory in these patients (Haematologica. 2019; 104(8):1580-1588; Am J Hematol. 2018; 93(12):1474-1484). Additional questions remain concerning the biological underpinnings of this population, but also, importantly, patterns of care and outcomes in the real-world setting. The objective of this study was to systematically evaluate patterns of care (diagnosis and management), as well as complications in AYA with MPN. Methods: Patients were recruited from the registry of the GQR LMC-NMP, an extensive provincial MPN network (&gt;11 academic and community centers across Quebec). AYA were age 18-40 years at diagnosis (control population, &gt; 40 years at diagnosis), diagnosed between 1978 and 2019 adhering to World Health Organization criteria, with exemption of bone marrow sampling in a subset. Standard risk stratification was according to MPN subtype. Conventional statistical methods were used for analyses (JMP® Pro 14.1.0 software; SAS Institute, Cary, NC, USA). Results: The AYA population consisted of 75 patients: n=15 (20%) polycythemia vera (PV), n=57 (76%) essential thrombocythemia (ET), and n=3 (4%) myelofibrosis (Table 1). Corresponding MPN subtypes in patients &gt; 40 years were 268 (40%) PV, 347 (51%) ET, and 63 (9%) MF. Median age at diagnosis was 34 years (range 18-40) in AYA vs 64 (41-95) in the older cohort. AYA were predominantly female (68% vs 55%; p=0.03). Median platelet count at diagnosis was higher in AYA vs non-AYA (641 vs 602 x10 9/L; p=0.08), with a greater proportion of patients (21% vs 10%; p=0.04) presenting with platelets &gt; 1000 x10 9/L. A trend towards more frequent palpable splenomegaly was observed in AYA (39% vs 25%; p=0.06). Driver mutation status in AYA vs older adults, respectively, was JAK2 (70% vs 87%) followed by CALR (28% vs 9%), MPL (2% in both), and triple negative (0% vs 2%) (p=0.001). Variant allele frequency (VAF) was significantly lower in AYA with median (range) of 20% (3-94) vs 39% (2-100) in non-AYA (p=0.02), with fewer AYA presenting VAF &gt; 20% at diagnosis (48% vs 76%; p=0.002). Risk stratification disclosed 24% of AYA (n=14) vs 53% (n=314) of non-AYA subjects to be high risk (p&lt;0.0001). Significantly fewer cardiovascular co-morbidities were reported in AYA (p&lt;0.003). Regarding work-up and management, markedly more AYA patients underwent bone marrow sampling vs older patients (65% vs 50%; p=0.01). While similar rates of phlebotomy and anti-platelet administration were reported (p=0.3 and 0.2, respectively), appreciably fewer AYA patients were treated with cytoreductive agents (41% vs 72% non-AYA, p&lt;0.0001). The nature of cytoreductive therapy varied between AYA vs older subgroups (p&lt;0.0001 for both hydroxyurea and interferon). A greater proportion of AYA (51%) vs non-AYA (9%) were also exposed to 2+ lines of cytoreduction (p&lt;0.0001). Similar rates of both arterial and venous thrombosis prior to/at diagnosis or post-diagnosis were found across AYA and non-AYA cohorts (p=0.9). Sites of predilection of venous events, however, were distinctive: AYA exhibited a preponderance of splanchnic vein and upper extremity thrombosis (72% and 14% respectively) vs non-AYA (20% and 7%) (p=0.03). Rates of hemorrhagic complications were significantly higher in AYA vs older adults (28% vs 16%; p=0.01), with distinctive patterns of bleeding, dominated by post-procedural/trauma/post-partum (24%) and mucocutaneous (52%) in AYA vs non-AYA (p=0.008). Kaplan-Meier survival analyses revealed proportional rates of thrombosis and hemorrhage-free survival but longer myelofibrosis-free (p=0.004) and overall survival (p=0.0006) in AYA vs older adults (Figure 1). Conclusions: AYA with MPN not only display unique clinical features but are also subject to distinct diagnostic and management practices. More rigorous investigational approaches and a propensity to address AYA with intensive cytoreductive strategies may reflect a signal of overtreatment of this population. Further, the incidence and nature of thrombotic/bleeding complications show distinctive patterns in AYA, requiring targeted vigilance and an individualized approach to monitoring and therapy. Figure 1 Figure 1. Disclosures Szuber: Novartis: Honoraria.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4840-4840
Author(s):  
Elena E. Khodunova ◽  
Elena N. Parovichnikova ◽  
Irina V. Galtzeva ◽  
Sergey M. Kulikov ◽  
Valentin G. Isaev ◽  
...  

Abstract Abstract 4840 The causes of drug resistance in acute leukemias (AL) have been studied very intensively and the key research was done on Bcl-2 family proteins. Last studies have showed that high level Bcl-2 expression in acute leukemia is really associated with drug resistance andpoor prognosis [Haematologica 2007, U. Testa]. It was demonstrated that lower Bax/Bcl-2 ratio (<0,3) was associated with FAB M0-M1 classes (p=.00001), poor-risk cytogenetics and poor prognosis [Blood 2003, G. Poeta]. But there were no studies on the dynamic evaluation of Bcl2 and Bax expression on CD34+ cells during chemotherapy. Renin-angiotensin system and angiotensin concertin enzyme (ACE) influence on leukogenesis is extensively investigated. It was reported that ACE expression on blast cells is high [Leuk Lymphoma 2006, S. Aksu]. Recent publications indicate that primitive hematopoietic precursors have different characteristics regarding ACE: CD34+ACE+cells transplanted into NOD/SCID mice contribute 10-fold higher numbers of multilineage blood cells than their CD34+ACE- counterparts and contain a significantly higher incidence of SCID-repopulating cells than the unfractionated CD34+ population [Blood 2008, V. Jokubaitis]. But it's still unknown how CD34+ACE+ cells in AL behave on and after chemotherapy. We have studied the dynamics of Bcl-2 and Bax expression by flow cytometry in CD34+ cells of peripheral blood (PB) and bone marrow (BM) in pts with AL. PB and BM samples were collected before treatment, on days +8, +36, only PB - on day + 21. Bcl-2 and Bax were detected on CD34+ cells by flow cytometry using specific monoclonal antibodies: CD34 (8G12, BD), Bcl-2 (100, BD), Bax (2D2, Santa Cruz). ACE (9B9, BD) expression was also evaluated. We calculated 10 000 cells in each sample. 10 pts were included in the study: 4 AML, 6 ALL. The control group comprised 4 healthy donors. At time of diagnosis CD34+ cells number in BM was 38,7%± 9,75, in PB - 38,3%± 8,14 in AL pts, not differing much in AML and ALL, and indicating blast cells population. CD34+ cells numbers in BM and PB of healthy donors were 1,35% and 0,23%, respectively. After induction therapy and WBC recovery (days +36-38) CD34+ cells number in AL pts decreased dramatically in BM to 3,83%±1,51 (p=0,001) and in PB to 0,98%± 0,29 (p=0,0001), indicating the efficacy of chemotherapy. The dynamics of Bcl-2, Bax and ACE expression on CD34+ cells of BM and PB in AL pts are presented in fig.1-6 As seen in the fig.1,2 CD34/Bcl-2 expression in BM is significantly higher (p=0,04) and in PB is similar in AL pts at the diagnosis comparing with donors. It's also worth to note that BM and PB CD34+ cells in donors had different expression characteristics of Bcl-2 demonstrating much higher level of antiapoptotic marker in PB cells. On the contrast CD34+ AL cells in BM and PB had similar characteristics regarding CD34/Bcl-2 expression. This expression level decreased substantially in BM at day +36 comparing with day 0 (p=0,04), but it never reached the donors level remaining extremely high and supposing the persistence of antiapoptotic activity in CD34+ cells in AL pts. It did not change at all during chemotherapy in PB cells, being identical to donors characteristics. The fig.2,3 demonstrate that, CD34/Bax expression in BM is almost 3-times higher (p=0,14) and in PB is twice lower (p=0,02) in AL pts in comparison with donors. It's interesting that CD34/Bax expression in leukemic BM and PB cells looks very similar, when in donors we had very low expression in BM and high - in PB. This fact demonstrates the heterogeneity of donor CD34+cells in BM and PB and points that leukemia CD34+cells in BM and PB are rather similar in Bax expression. Chemotherapy caused the significant augmentation of CD34/Bax expression in PB on day +8 (p=0,01) and near significant on day +21 (p= 0,09) showing the increased level of “dieing” cells in PB after cytostatic influence. The fig. 5,6 show that CD34/ACE coexpression in BM cells of AL pts and donors did not differ much at any time of evaluation. But CD34/ACE expression in PB cells of AL pts was much lower (p=0,02) than in donors and substantially increased at day +36 almost reaching the donor level. We may conclude that Bcl-2, Bax, ACE expression on CD34+ cells in AL pts and donors significantly differs, the dynamics of expression in AL while chemotherapy shows critical changes in CD34/Bcl-2 expression in BM, CD34/Bax and CD34/ACE in PB. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1593-1593
Author(s):  
Rabin R. Rabin ◽  
M. Monica Gramatges ◽  
Michael J. Borowitz ◽  
Shana L Palla ◽  
Xiaodong Shi ◽  
...  

Abstract Abstract 1593 Poster Board I-619 Many studies have now demonstrated the prognostic utility of measuring lymphocyte counts before, during and after non-transplant therapy in a wide range of malignancies. In children and young adults with acute lymphoblastic leukemia (ALL), low absolute lymphocyte counts (ALC) during induction chemotherapy are a strong predictor of adverse outcome, however the role of ALC in the era of minimal residual disease (MRD) is not known. We reviewed 171 pediatric patients with de novo ALL, age 1-21 years, treated at two institutions on the Children's Oncology Group P9900 series of trials from 2000-2006. Variables analyzed included ALC at diagnosis and days 15, 22 and 29 of induction chemotherapy; and other features including age, cytogenetics, WBC, absolute neutrophil count (ANC), platelets, therapeutic regimen, and minimal residual disease (MRD) status at day 29 (MRD-29). ALC at each time-point was evaluated for prognostic ability by univariate and multivariate analysis. Cut-point determined by Martingale residual analysis. We found high ALC at induction day 29 (ALC-29) to be predictive of improved relapse-free survival (EFS) and overall survival (OS), with a cut-point of 1500 cells/mL demonstrating the greatest predictive power. Patients with ALC-29 > versus <1500 had a 5-year EFS of 83% versus 66% (hazard ratio (HR) 2.2, p=0.018) and a 5-year OS of 96% versus 79% (HR 7.0, p=0.001) respectively. When compared directly to MRD-29 in multivariate analysis, ALC-29 demonstrated independent prognostic significance in EFS (HR 2.4, p=0.017) with striking HR/p-value for OS compared to MRD-29 (HR 10.9 vs. 4.9 and p=0.002 vs. 0.007). Importantly, ALC-29 also remained significant in multi-parameter multivariate analysis which included MRD-29 >0.01%, age >10y, WBC >50k, and favorable/unfavorable cytogenetics (HR 6.8, p=0.022). We found that ALC-29 discriminated subgroups with distinct differences in outcome among patients categorized by MRD-29. Among patients with MRD-29 <0.01%, those with ALC-29 <1500 had an 75/89% EFS/OS while those with ALC-29 >1500 had an excellent EFS/OS of 88/99% (HR 2.7/13.2, p=0.038/0.021). Most strikingly, among patients who were MRD positive (MRD-29 >0.01%), those with ALC-29 <1500 had a dismal EFS/OS of 33/41% versus those with ALC-29 >1500 who had a markedly superior EFS/OS of 69/92% (HR 2.0/9.1, p=0.24/0.041), providing clinically meaningful differences of 36% EFS and 51% OS. At 7 years the combination of ALC-29 and MRD-29 identifies three prognostic groups: a favorable group containing 60% of patients with EFS/OS 86/99% (MRDneg/ALChi), an intermediate group containing 32% of patients with EFS/OS 59-69/81-92% (MRDpos/ALChi and MRDneg/ALClo), and a very poor prognosis group containing 8% of patients with EFS/OS 33/41% (MRDpos/ALClo). These results suggest that ALC, a simple and readily obtainable test, constitutes a novel and powerful prognostic factor in children and young adults with ALL. ALC has prognostic significance that is independent of MRD and other known risk factors, and may refine MRD-based risk stratification, notably with the ability to identify a subset of MRD-positive patients with a good prognosis. Moreover, the association of higher ALC with improved outcome suggests an important role for host immune surveillance in mediating survival in childhood ALL. Disclosures No relevant conflicts of interest to declare.


PEDIATRICS ◽  
1970 ◽  
Vol 45 (2) ◽  
pp. 338-339
Author(s):  
F. Leef ◽  
G. Kende ◽  
B. Ramot

The paper by Dr. Finklestein and others,1 leaves one with the impression that testicular involvement in acute leukemia, an almost general finding at post mortem, is an uncommon clinical problem. We wonder whether lack of awareness might not be responsible for this, as our own experience suggests. In a clinic that follows at any time about 20 children and young adults with acute leukemia, we had seen only one case of testicular involvement (presenting symptom) in 3 years.


Blood ◽  
1995 ◽  
Vol 86 (1) ◽  
pp. 60-65 ◽  
Author(s):  
JT Holden ◽  
RB Geller ◽  
DC Farhi ◽  
HK Holland ◽  
LL Stempora ◽  
...  

Thy-1 (CDw90) is a phosphatidylinositol-anchored cell surface molecule which, when coexpressed with CD34 in normal human bone marrow, identifies a population of immature cells that includes putative hematopoietic stem cells. To date, the characterization of Thy-1 expression has been confined largely to normal tissues and cell lines. In this study, we evaluated the frequency and intensity of Thy-1 expression as defined by reactivity with the anti-Thy-1 antibody 5E10 in 38 cases of CD34+ acute leukemia (21 acute myelogenous leukemia [AML], 8 chronic myelogenous leukemia [CML] in blast crisis, and 9 acute lymphoblastic leukemia [ALL]). In 34 of 38 cases (89%) the CD34+ cells lacked expression of the Thy-1 antigen. High-density Thy-1 expression was found in 1 case of CML in lymphoid blast crisis, and low- density Thy-1 expression was identified on a portion of the leukemic cells in 2 cases of AML with myelodysplastic features, and 1 case of CML in myeloid blast crisis, suggesting a possible correlation between Thy-1 expression and certain instances of stem cell disorders such as CML and AML with dysplastic features. In contrast, the dissociation of Thy-1 and CD34 expression in the majority of acute leukemias studied suggests that the development of these leukemias occurs at a later stage than the hematopoietic stem cell. Characterization of Thy-1 expression in acute leukemia may eventually provide insights into the origin of the disease. In addition, separation of leukemic blasts from normal stem cells based on Thy-1 expression may prove useful in assessing residual disease, as well as in excluding leukemic blasts from stem cell preparations destined for autologous bone marrow or peripheral stem cell transplantation.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e18005-e18005
Author(s):  
Ali El Sayed ◽  
Patrick Michael Dillon ◽  
John Densmore ◽  
James Patrie ◽  
Hanna Kelly Sanoff

e18005 Background: Anthracyclines, the backbone of acute leukemia induction regimens, are associated with risk of cardiotoxicity. Routine cardiac imaging is routinely performed before induction treatment with anthracycline based regimens (ABR).There is no standard guideline on the optimal method of LVEF assessment, nor consensus for the treatment implication of a borderline LVEF finding. The NCCN recommends cardiac imaging for patients with prior heart disease, prior anthracycline use, or clinical symptoms of cardiac dysfunction. We examine the role of routine pre-induction cardiac imaging in young adults diagnosed with acute leukemia. Methods: A database search at the University of Virginia identified patients newly diagnosed with ALL and AML (including APML), between the ages of 18 and 49 from 2000 to 2011. Baseline demographics, pre-treatment cardiac imaging, and the induction chemotherapy regimen chosen were abstracted. Results: A total of 71 patients with ALL or AML were identified. Thirty-three patients (46.5%) were female, and the average age was 36.1 (SD=9.9 years, range 18 to 49 ). Median baseline LVEF was 62.5 (range 54-74). Out of 71 patients, 25 had a borderline LVEF between 50% and 60%.The lowest baseline EF was 54%. Among 25 patients with borderline LVEF, all received ABR for induction. Conclusions: In patients under age 50 with newly diagnosed acute leukemias, pre-treatment cardiac imaging did not alter standard induction therapy with ABR. This suggests that induction therapy should not be delayed to await baseline LVEF assessment. A potential cost saving may be achieved in this population. Routine cardiac imaging may be more helpful in older adults or patients with prior cardiac history or anticipated need for consolidation with an ABR (ie APML).


2016 ◽  
Vol 38 (2) ◽  
pp. 97-101 ◽  
Author(s):  
Esther P. Soundar ◽  
David Berger ◽  
Andrea Marcogliese ◽  
Lakshmi Srivaths

Author(s):  
Prasoon Sebastian ◽  
Abdul Majeed Kuruvadangal ◽  
Hitha Babu

Background: Acute leukemias are treated with intensive chemotherapy protocols which are associated with increased risk of infections. The objective of this study was to determine the incidence of febrile neutropenia and sepsis in acute leukemia patients during induction chemotherapy.Methods: In this prospective study we analysed the data of febrile neutropenia of forty-four patients of acute leukemia treated with intensive chemotherapy protocols. Study was conducted in hemato-oncology unit of Government Medical College, Kozhikode from January 2018 to December 2018. Events of the first month of induction were assessed, data entered in Microsoft excel and analysed with SPSS software.Results: Febrile neutropenia developed in all patients with AML induction therapy and 21.4% patients with ALL induction therapy. Causative organism was identified in 41.6% of febrile neutropenia episodes. Major focus of infection was lower respiratory tract followed by gastrointestinal tract. Fungal infection was identified in 6.8% cases. Mortality in AML induction was 31% and that of ALL induction was 3.57%. Infection was the most common cause of mortality. No clinical or laboratory parameters were found significant to predict outcome during induction chemotherapy in acute leukemia.Conclusions: Neutropenic fever and sepsis are the major cause of mortality in acute leukemia during induction chemotherapy. Early initiation of appropriate antibiotics will help to improve outcome in the treatment of leukemia.


Sign in / Sign up

Export Citation Format

Share Document