Low Expression of p53 Protein in Bone Marrow after Hematopoietic Stem Cell Transplantation Indicates Risk of Relapse in Pediatric Patients with Acute Lymphoblastic Leukemia

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4996-4996
Author(s):  
Kristin Mattsson ◽  
Emma Honkaniemi ◽  
Birgitta Sander ◽  
Britt Gustafsson

Abstract Advances in treatment over the last decades have resulted in almost 90% 5-year survival of children with acute lymphoblastic leukemia (ALL). Nonetheless, a minor group with unfavorable prognosis becomes candidates for hematopoietic stem cell transplantation (HSCT). After HSCT, blast count, minimal residual disease (MRD) and chimerism analysis are used as markers for successful engraftment and risk of relapse. However, additional markers are needed to complement the existing ones, as 30 % of ALL-patients still experience relapse after HSCT. Mutations in the tumor suppressor gene TP53 are detected in 2% at diagnosis of pediatric ALLs, but in 11-28 % after relapse. The aim of this study was to evaluate if alterations in the expressions of the TP53 encoded protein p53, analyzed by immunohistochemistry, could act as a prognostic marker in pediatric ALL patients post HSCT. Paraffin-embedded bone marrow samples were collected retrospectively from 49 children diagnosed with pre-B ALL and, treated with HSCT at Karolinska University Hospital between 1997-2010. Patients samples were requested from six different times during the course of the disease; time of diagnosis, before HSCT and at routine controls after HSCT at 0-3, >3-6, >6-12 and >12-24 months. For no patient data was available from all time periods. In total 176 samples were collected. A control group consisted of fifty-five children, where a bone marrow biopsy had been performed due to suspected bone marrow disorder, but a malignant blood disease had been excluded. Tissue micro array (TMA) prepared microscope slides were stained for the p53 protein using a monoclonal mouse anti-human antibody (clone DO-7, Leica), which detects both the wild-type and mutant forms of p53. Protein expression was examined by two independent examiners in a light microscope at high magnification (40X). Cells positive for the p53-protein identified by brown nuclear staining, were counted and calculated as a percentage of total cells per sample for each antibody respectively. Mean number of cells counted per sample was 695. Some samples were lost prior to final analysis due to either; sample core lost during TMA (n=12) or, sample had <100 cells which was minimum number of cells for inclusion in analysis (n=10). The final analysis included 154 bone marrow samples from 49 patients and 55 control samples. Data was analyzed in Statsoft Statistica 12.7. Nonlinear logistic regression was used to evaluate predictive value of the protein. Data was analyzed independently at each of the following times; diagnosis, time of HSCT and at 0-3, >3-6, >6-12 and >12-24 months post HSCT. A p value < 0.05 was considered significant. Interestingly, we found that a low percentage of cells positive for p53 protein expression in the bone marrow at >3-6 months after HSCT was associated with increased risk of relapse in pediatric ALL patients, odds ratio 0.82 (confidence interval 0.68-0.99) p= 0.041. Figure I shows the range of p53 expression in the two groups individually at each time. At >3-6 months after HSCT, the cut off value was calculated to 3.3% p53 positive cells, and was correct for 16 of 20 non-relapse cases and for 9 of 14 relapse cases (overall 73.5% correct). All relapses occurred between 6-50 months after HSCT (mean=20, median=14). The lower amount of p53 positive cells in the relapsed patients compared to the non-relapsed patients could possibly be due to an underlying mutation in TP53 that has altered the ability of producing a protein. This would be in similarity with the literature where it is suggested that TP53 mutations are more common at relapse in pediatric ALL patients. In summary, a lower expression of p53 protein in the bone marrow at >3-6 months after HSCT was significantly associated with increased risk of relapse. Evaluation of p53 protein expression by immunohistochemistry, in bone marrow from pediatric ALL-patients that undergo HSCT may be a potential additional marker for predicting relapse. Figure 1. Range and median percentage of p53 positive cells at different times in bone marrow from children with pre-B ALL. At >3-6 months after HSCT a higher quantity of p53 positive cells indicated a relapse free prognosis. Figure 1. Range and median percentage of p53 positive cells at different times in bone marrow from children with pre-B ALL. At >3-6 months after HSCT a higher quantity of p53 positive cells indicated a relapse free prognosis. Figure 2. Figure 2. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
1992 ◽  
Vol 79 (8) ◽  
pp. 1982-1986 ◽  
Author(s):  
CI Rivas ◽  
D Wisniewski ◽  
A Strife ◽  
A Perez ◽  
C Lambek ◽  
...  

Abstract Previous studies by others using metabolic labeling, cell lysis, and immunoprecipitation have reported elevated levels of p53 protein in blast cells derived from patients with acute lymphoblastic leukemia (ALL) and acute myeloblastic leukemia (AML), whereas p53 protein was not detected in normal light-density bone marrow cells. In this report, using the same detection methods, we confirm the negligible expression of p53 protein in normal light density marrow cells. However, we find clearly significant levels of p53 protein expression in enriched normal human marrow blast populations. Furthermore, using a panel of p53 specific monoclonal antibodies, we find the p53 protein constitutively synthesized by normal marrow blasts has the immunologic phenotype identified by PAb240 that reportedly recognizes a common conformational- dependent epitope on mutant p53. We have also found that the p53 immunologic subclass identified by PAb240 exists in normal human circulating lymphocytes either resting, serum starved, or PHA activated. In summary, it is clear that (1) normal marrow blast populations provide the appropriate control for assessing the levels of p53 protein expression in leukemic blast cells; and (2) PAb240 cannot be used to distinguish p53 mutated at the DNA level from normal p53 in fresh human hematopoietic cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 322-322
Author(s):  
Jean-Michel Cayuela ◽  
Paola Ballerini ◽  
Marina Romeo ◽  
Vahid Asnafi ◽  
Marie-Francoise Auclerc ◽  
...  

Abstract TEL-AML1 fusion transcripts are found in 25% of children with B-cell precursor ALL (BCP-ALL). From June 1993 to December 1999, 1195 children with BCP-ALL were included in the FRALLE 93 protocol. Out of these, 792 were evaluated for TEL-AML1 transcript expression. There is no difference in terms of initial features, DFS, EFS, survival between evaluated (792) and non evaluated (403) patients. Out of the 792 pts, 191 (24%) expressed TEL-AML1 transcripts at diagnosis. To assess the potential prognostic value of TEL-AML1 transcripts quantification, we have retrospectively analysed follow up marrow samples using Europe Against Cancer procedures for real time quantitative RT-PCR assay, on ABI PRISM 7700 (2 reference labs) and Light Cycler apparatus (1 reference lab). Out of the 191 TEL-AML1+ve pts, 83 were evaluated for MRD at different time points after induction therapy (median = D41 (34–55) (53 evaluable pts), at D111 (62–158) (62 pts), at D216 (159–325) (33 pts) and at D838 (365–1287) (49 pts). According to normalized Ct values, samples were attributed to 4 MRD level ranging from 0 to 3 and defined as follows: 0: Ct>40 ; 1 : 36<Ct≤40 ; 2 : 33<Ct≤36 ; 3 : Ct≤33, corresponding respectively to undetectable MRD ; MRD<10-4 ; 10-4≤MRD<10-3 ; MRD≥ 10-3, with respect to dilution of REH cDNA. Distribution of pts according to MRD level at different time points after induction treatment are summarized in the following table. Seventeen relapses have occurred at a median time of 41 months (17–73)(bone marrow: 7, BM + other: 5, testis: 3, CNS: 2). A level 2 positivity at the end of induction was associated with an increased risk of relapse of 3.31(95%CI:1.02 – 10.76, p =.047) while level 3 positivity was associated with a relative risk of 9.52 (95%CI: 2.91 – 31.08, p =.0002). Positivity at D111 was associated with an increased risk of relapse of 8.6 (2.0 – 38.5, p = 0.0042), whatever the level. Combination of data obtained at D41 and D111 allows to distinguish 3 subsets of pts with decreasing relapse-free survival: from 97.5% (95%CI: 85–100%) in pts with no positivity at D111 whatever the D41 result, to 75% (95%CI: 58–92%) in pts with MRD +ve at D111 with low level at D41 and 42% (95%CI: 14–69%) in pts with MRD +ve at D111 with level 2 or 3 at D41 (p<.0001). No other prognostic factor was found (age, sex, WBC, D8 steroid response, D21 bone marrow response) which renders the MRD profile unique in this matter. Conclusion: RQ-PCR-based MRD detection is a powerful prognostic tool in TEL-AML1+ve leukemia. Combination of two time points allows a relevant stratification of pts according to the risk of relapse, compatible with clinical decision making towards intensification or deescalation in the setting of controlled trials FU time point Number of pts in MRD classes (number of relapses) 0 1 2 3 Not evaluated D41 27 (3) 11 (2) 10 (4) 5 (4) 30 (4) D111 40 (2) 14 (6) 7 (2) 1 (1) 21 (6) D216 29 (2) 2 (1) 1 (0) 1 (1) 50 (13) D838 47 (8) 1 (0) 1 (1) 0 34 (8)


Blood ◽  
1992 ◽  
Vol 79 (8) ◽  
pp. 1982-1986 ◽  
Author(s):  
CI Rivas ◽  
D Wisniewski ◽  
A Strife ◽  
A Perez ◽  
C Lambek ◽  
...  

Previous studies by others using metabolic labeling, cell lysis, and immunoprecipitation have reported elevated levels of p53 protein in blast cells derived from patients with acute lymphoblastic leukemia (ALL) and acute myeloblastic leukemia (AML), whereas p53 protein was not detected in normal light-density bone marrow cells. In this report, using the same detection methods, we confirm the negligible expression of p53 protein in normal light density marrow cells. However, we find clearly significant levels of p53 protein expression in enriched normal human marrow blast populations. Furthermore, using a panel of p53 specific monoclonal antibodies, we find the p53 protein constitutively synthesized by normal marrow blasts has the immunologic phenotype identified by PAb240 that reportedly recognizes a common conformational- dependent epitope on mutant p53. We have also found that the p53 immunologic subclass identified by PAb240 exists in normal human circulating lymphocytes either resting, serum starved, or PHA activated. In summary, it is clear that (1) normal marrow blast populations provide the appropriate control for assessing the levels of p53 protein expression in leukemic blast cells; and (2) PAb240 cannot be used to distinguish p53 mutated at the DNA level from normal p53 in fresh human hematopoietic cells.


Gut ◽  
2012 ◽  
Vol 62 (12) ◽  
pp. 1676-1683 ◽  
Author(s):  
Florine Kastelein ◽  
Katharina Biermann ◽  
Ewout W Steyerberg ◽  
Joanne Verheij ◽  
Marit Kalisvaart ◽  
...  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1432-1432
Author(s):  
Christopher Daniel Chien ◽  
Sang Minh Nguyen ◽  
Haiying Qin ◽  
Elad Jacoby ◽  
Terry J. Fry

Abstract Despite 5-year survival rates nearing 90%, leukemia is the most frequent cause of death from cancer in children with treatment failure primarily caused by relapse after remission. Therefore, it is critical that we identify new therapies that specifically address leukemic persistence during therapy. A cohort of acute lymphoblastic leukemia (ALL) patients that have an increased risk of relapse and consequently poorer overall survival have leukemia that overexpresses cytokine receptor-like factor 2/thymic stromal lymphopoietin receptor (CRLF2/TSLPR). CRLF2/TSLPR overexpressing patients have rates of relapse nearly double the rate of non-overexpressing patients despite comparable remission rates to low-risk patients and account for half of the cases of high risk Philadelphia-like ALL. We hypothesize that TSLP/TSLPR axis may promote relapse of ALL through overexpressed TSLPR, which sensitizes leukemic blasts to low levels of TSLP in bone marrow (BM) niches promoting survival. To test this theory we generated a TSLPR overexpressing syngeneic murine leukemia (TSLPRhigh) through viral transduction of a transplantable pre-B cell ALL line (TSLPRlow). This TSLPRhigh leukemia has comparable TSLPR expression levels to what is found on human ALL that overexpress TSLPR and the transduced TSLPR is functional with increased phosphorylation of STAT5 protein in response to TSLP stimulation. When ALL lines were injected into immunocompetent mice, we observed an 8 fold difference in the percentage of TSLPRhigh vs. TSLPRlow ALL in the BM 5 days after injection, when leukemia accounted for less than 5% of the BM corresponding to an early stage of ALL progression. Interestingly, in vitro and in vivo cell growth and late-stage lethality were no different between the TSLPRhigh and TLSPRlow ALL indicating that TSLP does not alter ALL proliferation. From this data we can infer that TSLP/TSLPR signaling is likely most critical at early stages of leukemia development when BM stromal niches are intact. Hypothesizing that these niches are the source of TSLP ligand we sought to identify the cells responsible for secreting TSLP in the BM. Indeed, basal levels of TSLP mRNA are present in the BM, but the expression is quite low. Interestingly, TSLP mRNA and protein were markedly induced in murine BM stromal cell lines and primary BMSCs by the inflammatory cytokines IL-1a and TNF-a as has previously been reported for other types of TSLP-producing human cells. Since it is well established that cytotoxic chemotherapeutics can mediate an inflammatory response in patients, we investigated whether cytotoxic agents can cause release of inflammatory cytokines from BM cell populations. We treated primary murine BMSCs, BM hematopoietic cells, and ALL lines with low doses of chemotherapy and observed that the pyrimidine analog cytarabine (Ara-C) was particularly potent in upregulating IL-1a expression from BM hematopoietic cells and ALL and not in BMSCs suggesting that inflammatory cytokine release by hematopoietic cells may induce TSLP production by BMSCs. We set out to determine if IL-1a administration to mice could enhance the early progression of ALL in vivo. Indeed, we found that TSLP mRNA and protein were elevated in the BM and serum of mice injected with IL-1a respectively and that there was significant increase in the percentage of early ALL infiltration in the BM of mice bearing TSLPRhigh ALL but not TSLPRlow ALL suggesting a dependence on high levels of expression of TSLPR to respond to IL-1a -induced TSLP production. In addition, we confirmed the necessity of TSLPR-driven early ALL progression on the presence of the TSLP ligand as we observed no significant increase in TSLPRhigh ALL in TSLP deficient mice after stimulation with IL-1a. Furthermore, the early increase in TSLPRhigh leukemia burden can be reversed by using a TSLP blocking antibody demonstrating that targeting the TSLP/TSLPR axis may be therapeutically relevant. These data demonstrate that TSLP secreted in the BM induced by inflammatory cytokines such as IL-1a can drive accelerated early progression of ALL. These inflammatory cytokines can be induced by cytotoxic agents suggesting that chemotherapy can indirectly provide an unintended advantage to TSLPR overexpressing ALL. Finally, we postulate that therapies targeting the TSLP signaling axis would decrease the risk of relapse in TSLPRhigh ALL particularly in the context of standard therapy. Figure 1. Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2261-2261
Author(s):  
Marissa den Hoed ◽  
S.M.F. Pluijm ◽  
Hester A. de Groot-Kruseman ◽  
Mariël L. Te Winkel ◽  
Erica L.T. van den Akker ◽  
...  

Abstract Background: Body mass index (BMI: kg/m2) and change in BMI during treatment might influence treatment outcome of pediatric patients with acute lymphoblastic leukemia (ALL). However, previous studies in pediatric acute lymphoblastic leukemia reported contradictory results. Therefore, we studiedthe influence of (change in) BMI on treatment outcome in pediatric ALL patients who were treated according to a dexamethasone-based protocol (Dutch Childhood Oncology Group [DCOG] ALL-9). Patients and Methods: Data on body composition were prospectively collected from a cohort of newly diagnosed Dutch pediatric ALL patients (N=762, age 2-17 years), treated from 1997-2004. BMI at diagnosis was expressed as standard deviation scores (SDS) and categorized into underweight (≤–1.8SDS), or normal weight and overweight (>–1.8SDS). BMI loss was defined after 32 weeks of treatment. Dual X-ray absorptiometry scans were performed in a nested single center cohort (n=106) to assess the contribution of %fat and lean body mass to BMI. Multivariate analyses were corrected for age at diagnosis and risk treatment grpup. Results: Multivariate analyses showed that patients with underweight (8%) had an increased risk of relapse (Hazard Ratio (HR) 1.79, 95% CI: [1.04-3.10], and a similar overall survival (HR 1.10 [0.57-2.10]). BMI loss during the first 32 weeks of treatment was associated with a decreased overall survival (HR 2.10 [1.14-3.87]), and a similar risk of relapse (HR 1.27 [0.70-2.30]) compared to patients without BMI loss. Dual X-ray absorptiometry revealed that BMI loss mainly consisted of a loss of lean body mass and gain in %fat. Conclusion: Underweight at diagnosis is associated with an increased risk of relapse and a BMI loss early during treatment is associated with an increased mortality. This suggests that these patients might benefit from exercise interventions and a high-quality nutrient diet during therapy. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document