P53 Protein Overexpression By Immunohistochemical Staining Is Correlated with TP53 Mutation Burden and Adverse Clinical Outcome in Myelodysplastic Syndromes

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4121-4121 ◽  
Author(s):  
Nguyen Johnny ◽  
Kathy L McGraw ◽  
Rami S. Komrokji ◽  
David Sallman ◽  
Najla H. Al Ali ◽  
...  

Abstract Background: TP53 mutations have been reported in 5-10% of patients with myelodysplastic syndromes (MDS) with a higher frequency in those who harbor del(5q) and complex cytogenetics. Next generation sequencing (NGS) data in large clinical cohorts has revealed TP53 mutation is a strong independent prognostic factor. Limited studies have shown an association of p53 protein overexpression with TP53 mutation as well as other clinical characteristics, such as blast count, cytogenetics, and mutant TP53 variant allele frequency (VAF). The study aims to validate the association of p53 overexpression by immunohistochemical (IHC) staining with TP53 mutation, and to explore its relationship to clinical outcome and mutation burden in MDS. Methods: We retrospectively analyzed MDS patients with or without transformation to acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) diagnosed between 7/2013 and 5/2015. Only those cases with available bone marrow (BM) biopsies with good quality (>1.0 cm in length) corresponding to the NGS testing date were included. Five normal BMs were used as controls. IHC was performed according to our institutional standard protocol and modified in accordance with the manufacturer. Nuclear expression of p53 was assessed semi-quantitatively using an IHC score calculated by multiplying IHC stain intensity with percent positivity in hematopoietic cells. Paired t-test was used for numerical parameters. Hazard ratios were generated using the standard cox proportional harzard model. Survival was analyzed by the Kaplan-Meier method and overall survival (OS) was defined as the duration between date of diagnosis and date of death. Results: Of 201 patients with myeloid malignancies, 29 (14%) harbored clinically significant mutations by NGS testing. Twenty two of these (9 AML-MRC,13 MDS) had available BM biopsies. An additional 32 patients (27 MDS, 5 AML) with wild type (WT) TP53 were included. Clinicopathologic differences of these groups are summarized in Figure 1. IHC analysis showed significantly (p=1.21x10-6) elevated nuclear p53 expression in TP53 mutated patients (mean score, 1.11± 0.164) compared to WT (0.037 ±0.014). A higher p53 IHC score in mutated patients correlated with a complex karyotype (n=14) compared to those without (n=6) (1.235 ± 0.206, and 0.580 ± 0.257, respectively, p=0.054). A significant positive correlation between IHC score and cytogenetic risk group according to revised international prognostic scoring system ( R-IPSS) existed in our cohort (p<0.001). Importantly, p53 overexpression directly correlated with TP53 VAF within the MDS group (r=.855, p<0 .001). We found no correlation between p53 IHC score and blast count in mutated TP53 patients (r=0.355, p=0.110), but a significant correlation in those with WT TP53 (r=0.527, p=0.002), which merits further investigation. The proportion of del(17p) or del(5q) was greater in the TP53 mutated patients vs. WT patients [40% and 90% in mutated TP53 vs. 4% and 12% in WT, respectively). Median overall survival (OS) was 86 months (95%CI, 7-166) and median follow-up duration was 27 months (95% CI 7-46). The sensitivity and specificity of using a p53 IHC score in predicting TP53 mutation status using a cutoff of 1.00 was 59.1% and 100%, respectively. The sensitivity and specificity of using a p53 IHC score in predicting TP53 mutation status using a cutoff of 0.500 was 77.3% and 100%, respectively. Median OS was shorter in mutated patients [16 months (95% CI, 8-23)] compared to WT (not reached) (p=0.001). OS hazard ratio (HR) of mutant TP53 was 4.6 (95% CI, 1.8-12) (p= 0.002). We also found significant differences in OS by IHC score. Median OS was not reached in patients with an IHC score <1 compared to >1.0 [16 month (95% CI, 8-23)] (p= 0.007). Similarly, with a p53 IHC score cutoff of 0.5, median OS was again not reached in those patients with <0.5 IHC score but was 16 months (95% CI, 8-25) in those >0.5 (p=0.015). HR for IHC score > 0.5 was 3 (95% CI, 1.2-8), p=0.02. HR for IHC score >1.0 was 3.5 [(95% CI, 1.3-9), p=0.01]. Conclusion: p53 overexpression in MDS patients correlates with mutated TP53 and mutation burden. A higher IHC score was associated with poorer clinical features and outcomes. A larger cohort is warranted to define its diagnostic or prognostic utility. Figure 1. Clinicopathologic Characteristics in Our MDS/AML-MRC Cohort Figure 1. Clinicopathologic Characteristics in Our MDS/AML-MRC Cohort Disclosures Komrokji: Celgene: Consultancy, Research Funding; Pharmacylics: Speakers Bureau; Incyte: Consultancy; Novartis: Research Funding, Speakers Bureau. Padron:Incyte: Research Funding; Novartis: Speakers Bureau. Lancet:Celgene: Consultancy, Research Funding; Amgen: Consultancy; Pfizer: Consultancy; Boehringer-Ingelheim: Consultancy; Kalo-Bios: Consultancy; Seattle Genetics: Consultancy. List:Celgene Corporation: Honoraria, Research Funding.

2022 ◽  
Vol 29 ◽  
Author(s):  
Sebastian M. Klein ◽  
Maria Bozko ◽  
Astrid Toennießen ◽  
Nisar P. Malek ◽  
Przemyslaw Bozko

Background: Ovarian cancer is one of the most aggressive types of gynecologic cancers. Many patients have a relapse within two years after diagnosis and subsequent therapy. Among different genetic changes generally believed to be important for the development of cancer, TP53 is the most common mutation in the case of ovarian tumors. Objective: Our work aims to compare the outcomes of different comparisons based on the overall survival of ovarian cancer patients, determination of TP53 status, and amount of p53 protein in tumor tissues. Methods: We analyzed and compared a collective of 436 ovarian patient’s data. Extracted data include TP53 mutation status, p53 protein level, and information on the overall survival. Values for p53 protein level in dependence of TP53 mutation status were compared using the Independent-Samples t-Test. Survival analyses were displayed by Kaplan-Meier plots, using the log-rank test to check for statistical significance. Results: We have not found any statistically significant correlations between determination of TP53 status, amount of p53 protein in tumor tissues, and overall survival of ovarian cancer patients. Conclusion: In ovarian tumors both determination of TP53 status as well as p53 protein amount has only limited diagnostic importance.


2008 ◽  
Vol 139 (2_suppl) ◽  
pp. P42-P42
Author(s):  
Sohail M. Awan ◽  
Adnan Syed ◽  
Shehzad Ghaffar

Objective 1. To determine whether p53 protein expression is prognostic indicator of patient survival. 2. Learn its correlation with habits and histological variables of the tumor in Pakistani patients. Methods A retrospective case series of 140 patients with primary oral squamous cell carcinoma at a tertiary care setting during the period of January 1991 to Dec 2004. p53 protein overexpression was investigated by means of immunohistochemistry. Results of p53 overexpression were evaluated in relation to different clinicopathological parameters and survival. Overall survival and disease-free survival were measured. Event time distributions for these end points were estimated by use of the method of Kaplan and Meier and compared by use of the log-rank statistic. Factors associated with p53 status were selected on cross-tabulations and logistic regression. Cross-tabulations were analyzed by use of the chi-square test or Fisher's exact test, where appropriate. Results were summarized as odds ratio (OR) with corresponding 95% Confidence Interval. Results Overexpression of p53 protein was observed in 75 patients (54%). Patients with p53 negative tumors had improved overall survival (OS) when compared with patients with p53 positive tumors (p=0.036). The p53 overexpression did not correlate with disease-free survival (DFS) (p=0.126), histological differentiation (p=0.611) and patients’ habits (p=0.894) In univariate analysis, AJCC stage, nodal status, tumor size and histological grade were significantly associated with shortened OS and DFS. Conclusions Our study supports that patients with p53 overexpression had a significantly poor overall survival compared to p53 negative patients. However, p53 overexpression was not associated with patients disease-free survival.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4284-4284
Author(s):  
Blanka Kubesova ◽  
Sarka Pavlova ◽  
Jitka Malcikova ◽  
Jitka Kabathova ◽  
Lenka Radova ◽  
...  

Abstract Myeloproliferative neoplasms (MPN) are characterized by abnormal proliferation of myeloid lineages and a tendency toward leukemic transformation. Inactivation of tumor suppressor TP53 has been repeatedly associated with MPN transformation into secondary acute myeloid leukemia (sAML), but no fully expanded TP53 mutated clones in chronic phase of MPN were reported. The link between TP53 mutations and widely used cytoreductive treatment by hydroxyurea (HU) still remains controversial. To which extent TP53 mutations represent a risk of disease progression is not known. We aimed to search for low-burden TP53 mutated subclones in chronic-phase-MPN patients and to correlate presence of these clones with therapy, disease course, and other clinical features. In total we analyzed 220 patients by ultra-deep next generation sequencing (NGS). We detected TP53 mutations in 39 (18 %) patients with variant allelic frequency of 0.1-16.3 % at the first examination. The analysis of 136 patients treated with hydroxyurea or other drugs (anagrelide (ANA), interferon α (IFN)) for more than 4 years, as well as a group of 84 patients untreated by cytoreductive drugs, showed that TP53 mutations occurrence in chronic phase is independent of hydroxyurea use, disease type, and JAK2/CALR/MPL status. Mutations were found in 17/72 (24 %) HU treated patients, in 11/64 (17 %) patients treated by other drugs, and in 11/84 (13 %) untreated patients. Median size of mutated clones was 0.5 % and was not influenced by previous treatment. In 10 patients we found more than one mutation. In patients harboring TP53 mutations, retrospective samples were examined if available to explore the clonal evolution of TP53 mutated clones. The respective TP53 mutations were found in 13/20 cases analyzed; out of them, in 4 cases the mutation was found even in a diagnostic sample. Follow-up samples were examined in 28 patients with TP53 mutations and the mutation burden changed during the monitored time in majority of patients; however, the expansion into dominant clone was observed in one patient only. When all data from retrospective and prospective analyses taken together (30 patients), the median follow-up was 7.2 years. TP53 mutation burden tended to increase in 14 patients. In 6 patients the mutation burden remained stable and in 4 patients it fluctuated. In 6 patients the mutated clone size decreased; out of them in 2 originally very low-burden mutations (0.2 %) were not detectable in samples taken 15 and 3.4 years later. We did not observe any correlations of different patterns in TP53 mutation changes with therapy or other clinical characteristics (disease type, driver mutation, time from diagnosis, treatment response). Further, we assessed the TP53 mutation impact on overall survival and leukemic transformation. The mutations did not negatively affect disease progression or overall survival either from diagnosis or from mutation identification. sAML developed in 2 patients with TP53 mutations 17.9 (treated with IFN) and 8.3 (treated with HU) years from diagnosis. In the latter patient, the sAML developed from a different clone as it was TP53-wt, JAK2-wt, although 2 TP53 mutated clones within JAK2 mutated clone were detected in chronic phase. On the other hand, we have observed another interesting case where the TP53 mutation burden grew rapidly from 10 % up to nearly 100 % during the follow-up. In contrast to published data, this patient did not show any clinical signs of disease progression for 2 years after the expansion and died of MPN unrelated cause. In summary, using highly sensitive method we showed that low-burden TP53 mutations are present in MPN chronic phase. Neither their presence nor their size is associated with previous therapy and has impact on overall survival or leukemic transformation. Monitoring of TP53 mutations during the disease course showed that their clonal development is rather variable; nevertheless, TP53 minor mutations may represent a pool for future clonal evolution. Supported by MZ CR-RVO (FNBr, 65269705), MUNI/A/1028/2015, H2020 692298, MZO AZV 15-31834A, 15-30015A, MEYS LQ1601 and LM2015064. Disclosures Gisslinger: Baxalta: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; AOP Orphan: Consultancy, Honoraria. Mayer:AOP Orphan Pharmaceuticals: Research Funding; Novartis: Research Funding. Kralovics:AOP Orphan: Research Funding; Qiagen: Membership on an entity's Board of Directors or advisory committees.


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii32-iii32
Author(s):  
H Noor ◽  
R Rapkins ◽  
K McDonald

Abstract BACKGROUND Tumour Protein 53 (TP53) is a tumour suppressor gene that is mutated in at least 50% of human malignancies. The prevalence of TP53 mutation is much higher in astrocytomas with reports of up to 75% TP53 mutant cases. Rare cases of TP53 mutation also exist in oligodendroglial tumours (10–13%). P53 pathway is therefore an important factor in low-grade glioma tumorigenesis. Although the prognostic impact of TP53 mutations has been studied previously, no concrete concordance were reached between the studies. In this study, we investigated the prognostic effects of TP53 mutation in astrocytoma and oligodendroglioma. MATERIAL AND METHODS A cohort of 65 matched primary and recurrent fresh frozen tumours were sequenced to identify hotspot exons of TP53 mutation. Exons 1 to 10 were sequenced and pathogenic mutations were mostly predominant between Exons 4 and 8. The cohort was further expanded with 78 low grade glioma fresh frozen tissues and hotspot exons were sequenced. Selecting only the primary tumour from 65 matched tumours, a total of 50 Astrocytoma cases and 51 oligodendroglioma cases were analysed for prognostic effects of TP53. Only pathogenic TP53 mutations confirmed through COSMIC and NCBI databases were included in the over survival and progression-free survival analysis. RESULTS 62% (31/50) of astrocytomas and 16% (8/51) of oligodendrogliomas harboured pathogenic TP53 mutations. Pathogenic hotspot mutations in codon 273 (c.817 C>T and c.818 G>A) was prevalent in astrocytoma with 58% (18/31) of tumours with these mutations. TP53 mutation status was maintained between primary and recurrent tumours in 93% of cases. In astrocytoma, overall survival of TP53 mutant patients was longer compared to TP53 wild-type patients (p<0.01) but was not significant after adjusting for age, gender, grade and IDH1 mutation status. In contrast, astrocytoma patients with specific TP53 mutation in codon 273 showed significantly better survival compared to other TP53 mutant and TP53 wild-type patients combined (p<0.01) in our multivariate analysis. Time to first recurrence (progression-free survival) of TP53 mutant patients was significantly longer than TP53 wild-type patients (p<0.01) after adjustments were made, while TP53 mutation in codon 273 was not prognostic for progression-free survival. In oligodendroglioma patients, TP53 mutations did not significantly affect overall survival and progression-free survival. CONCLUSION In agreement with others, TP53 mutation is more prevalent in Astrocytoma and mutations in codon 273 are significantly associated with longer survival.


eLife ◽  
2016 ◽  
Vol 5 ◽  
Author(s):  
Gilles Gadea ◽  
Nikola Arsic ◽  
Kenneth Fernandes ◽  
Alexandra Diot ◽  
Sébastien M Joruiz ◽  
...  

TP53 is conventionally thought to prevent cancer formation and progression to metastasis, while mutant TP53 has transforming activities. However, in the clinic, TP53 mutation status does not accurately predict cancer progression. Here we report, based on clinical analysis corroborated with experimental data, that the p53 isoform Δ133p53β promotes cancer cell invasion, regardless of TP53 mutation status. Δ133p53β increases risk of cancer recurrence and death in breast cancer patients. Furthermore Δ133p53β is critical to define invasiveness in a panel of breast and colon cell lines, expressing WT or mutant TP53. Endogenous mutant Δ133p53β depletion prevents invasiveness without affecting mutant full-length p53 protein expression. Mechanistically WT and mutant Δ133p53β induces EMT. Our findings provide explanations to 2 long-lasting and important clinical conundrums: how WT TP53 can promote cancer cell invasion and reciprocally why mutant TP53 gene does not systematically induce cancer progression.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2076-2076
Author(s):  
Paloma Martin ◽  
Toon Min ◽  
Alison Morilla ◽  
Sarah Hockley ◽  
Ayoma D. Attygale ◽  
...  

Abstract B-cell chronic lymphocytic leukaemia (B-CLL) is characterized by a highly variable clinical course. TP53 abnormalities in B-CLL are strongly associated with resistance to purine analogues, short survival and large-cell transformation. Several methods can be used to investigate TP53 inactivation: FISH (gene deletion), immunohistochemistry -IHC- (protein overexpression), and DNA sequencing (mutations). The relationship between TP53 deletion and p53 protein overexpression is unclear, and it is uncertain which method is the best to assess TP53 dysfunction and which provides the most useful clinical information. In 92 CLL patients we have correlated the frequency of p53 protein overexpression by IHC, with TP53 deletion status by FISH and subsequently we have examined the response to Fludarabine in these patients. The male/female ratio was 1.35 (52M/40F), with a median age at diagnosis of 54 years (range 25–78 years). The distribution of clinical stage at diagnosis was as follows: 47% (43 cases) stage A, 16% (15 cases) stage A progressive, 14% (13 cases) stage B, 11% (10 cases) stage C, and unknown in 12% (11 cases). Fourteen cases (15%) were untreated and seventy-eight cases (85%) were previously treated. Among the latter, 66 (84%) received Fludarabine, and 9 (11%) received Campath-1H (as a first line of treatment in 5 patients). In all cases peripheral blood samples were used for FISH analysis and IHC was carried out on bone marrow biopsies. Cases were selected on the basis of whether FISH and IHC were analysed at the same time point. Deletions of TP53 (17p13) were detected by FISH (Vysis); a threshold of &gt;20% of cells with p53 deletion was considered clinically significant. For IHC detection a monoclonal antibody (clone BP53-12, Novocastra) was used. Cases were considered positive when p53 stained the majority of lymphocyte nuclei, those cases with few or weakly positive cells in proliferation centres were considered negative. Correlation with FISH (&gt;20% deleted cells) and IHC showed 69 cases (75%) negative for both assays and 6 cases (7%) positive for FISH and IHC. The remaining 17 cases (18%) showed discordant results; 6 cases showed TP53 deletion &gt;20% by FISH but showed no p53 overexpression by IHC, 4 cases had overexpression of p53 and no TP53 deletion, 3 cases had p53 overexpression with TP53 deletion between 10–20%, and 4 cases showed TP53 deletion between 10–20% but no overexpression of p53. This is compatible with a sensitivity of 50% and specificity of 91%. In order to understand the mechanism better, mutation analysis of TP53 gene will be done in discordant cases. All patients positive for both results and a minority of patients (16%) with no p53 abnormalities detected by FISH or IHC were refractory to Fludarabine. Within the discordant group, most of refractory patients (67%) were found in cases with p53 overexpression and TP53 deletion between 10–20%. In conclusion, the combination of both methods (FISH and IHC) may be useful to identify CLL cases with an adverse prognosis and non responders to Fludarabine. A large study with more cases is necessary to clarify the underlying mechanisms involved in those cases with p53 overexpression without TP53 deletion.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2427-2427 ◽  
Author(s):  
Thorsten Zenz ◽  
Raymonde Busch ◽  
Anna Fink ◽  
Dirk Winkler ◽  
Kirsten Fischer ◽  
...  

Abstract Abstract 2427 Introduction: The introduction of chemo-immunotherapy (R-FC) has led to impressive improvement of response, PFS and remarkably overall survival in CLL. Irrespective of these improvements, the genetic markers remain important predictors of outcome. With increasing intensity of 1st line treatment, the proportion of patients with refractory disease is decreasing. At the same time the selective pressure on tumor cells increases. Relatively little is known about the genetic profile of risk groups as defined by the length of 1st remission. We therefore analysed the CLL8 cohort (FC vs. FCR in untreated and fit patients with CLL) to identify the genetic profile (before 1st line treatment) of patients either not responding to (refractory) or relapsing early after first line therapy. Methods and material: In order to characterize the genetic profile of refractory CLL and CLL with early relapse we formed 4 cohorts based on response and response duration after initial therapy. We selected F-refractory (no PR/CR or PR/CR < 6 months), patients with PFS 6−<12 months, 12−<24 months and ≥ 24 months for the analysis. Median follow-up was 37.7 months. Genetic characterization was performed in a central laboratory (Ulm). Data was available for IGHV (n=587), TP53 mutation (n=592), and FISH (n=581). Results: Based on the above definition 84/767 (11%) patients were F-refractory. Very short PFS was observed in 43/767 (5.6%)(6−<12 months) and 110/767 (14.3%)(12−<24 months). The overall survival (OS) of patients in the 4 categories was significantly different with median OS (from study entry) of 21.9 months (F-refractory patients), 21.2 months (PFS 6−<12 months), and 47.3 months (12−<24 months) compared to not reached in patients with a median PFS ≥24 months. When comparing the treatment arms, the overall number of patients with short remission was lower in the FCR arm (Table 1). As shown in Table 1, the incidence of 17p- and TP53 mutation is highest in the F-refractory group with similar distribution in both treatment arms (34.4% 17p-; 43.8% TP53). In the subgroup of patients with very short PFS (6−<12 months) the incidence of highest risk genetic aberrations was still high (17p- 28.1%; TP53 mutation 23.5%). In contrast the fraction of patients with these aberrations was very low (1.5 and 4.1% resp.) in the group of patients with long PFS (≥24 months). Conversely, the incidence of good risk genetics (e.g. 13q- single) increased with length of remission (Table 1). Similarly, the incidence of trisomy 12 was higher in patients with longer PFS (11.3% and 11.4% for PFS 12−<24 months)(Table 1). Conclusion: Early relapse (within 24 months) is associated with high risk genetics (TP53 mutation and 17p deletion). In addition to patients with F-refractory CLL, patients relapsing within 6–12 and 12–24 months after intense 1st line treatment have a poor overall survival. Decisions on 2nd line treatment options should integrate genetic characterisation and remission duration. Disclosures: Zenz: Roche: Honoraria; Boehringer: Honoraria; GSK: Honoraria; Celgene: Honoraria. Fink: Roche: Travel grant. Fischer: Roche: Travel grant. Kneba: Roche: Honoraria, Research Funding. Boettcher: Roche: Research Funding. Mendila: Roche: Employment. Wenger: Roche: Employment. Hallek: Roche: Honoraria, Research Funding. Döhner: Roche: Research Funding. Stilgenbauer: Roche, Celgene, GSK, Boehringer, Genzyme: Honoraria, Research Funding.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1663-1663 ◽  
Author(s):  
Koichi Takahashi ◽  
Feng Wang ◽  
Seth Sahil ◽  
Jianhua Zhang ◽  
Curtis Gumbs ◽  
...  

Abstract Background: Recent advance in massively parallel sequencing technology has enabled identification of a number of novel somatic driver mutations in myelodysplastic syndromes (MDS). While these efforts have clearly advanced our understanding of MDS pathogenesis, clinical implication of driver mutations in MDS is less studied. Methods: We performed whole exome sequencing on bone marrow aspiration samples obtained from 114 consecutive patients with untreated MDS. Exome capture was performed using Agilent's SureSelect V4 and sequencing was conducted using Illumina's HighSeq 2000 platform. Sequencing achieved median 124x coverage for the targeted exons. Mutect and Pindel algorithm were used to call single nucleotide variants (SNV) and small indels against virtual common normal reference. Annotation of high-confidence driver mutations followed the previous publication by Pappaemmanuil et al. (Blood 2013). Clonal heterogeneity of driver mutations was assessed in patients who have 2 or more driver mutations by Pearson's goodness of fit test. Results: Among the 114 patients with MDS, total 221 high-confidence driver mutations were detected in 39 genes by sequencing. Eighty eight percent (100/114) of the patients were found to have at least one driver mutation. The number of driver mutation ranged from 1 to 5 per case. Commonly detected driver mutations include TET2 (25%), SRSF2 (22%), ASXL1 (20%), RUNX1 (19%), TP53 (12%), SF3B1 (9%), and U2AF1 (9%). As a rare driver mutation, we confirmed ETNK1 p. N244S mutation in 2 MDS patients (2%). This mutation was recently described as a recurrent somatic mutation in atypical CML (Gambacorti-Passerini et al. Blood 2015). Clonal heterogeneity of driver mutations was evaluable in 65 patients (57%). Nineteen patients were found to have clonal heterogeneity in driver mutations (29%). Among the 114 patients, 61 patients (54%) were treated with HMA therapy. Complete response (CR), partial response (PR), and hematological improvement (HI) was observed in 22 (36%) patients, 4 (7%) patients, and 5 (8%) patients, respectively. Presence of TET2 mutation did not predict response to HMA therapy in this series (P = 0.57) even when we restricted to TET2 mutations with variant allele frequency (VAF) >10%. There was a trend toward poor response to HMA therapy in ASXL1 mutated patients (P = 0.074). None of the other driver mutations were predictive of response to HMA therapy as a sole. However, patients who were found to carry 4 or more driver mutations had significantly poor response to HMA therapy (CR rate 0%) compared to patients with less than 4 driver mutations (P = 0.035). Presence of clonal heterogeneity in driver mutations was not predictive of response to HMA therapy (P = 0.43) In regards to survival outcome, presence of SF3B1 mutation predicted favorable overall survival (OS, P = 0.02) while TP53, and DNMT3A mutations were associated with worse OS (P < 0.001 and P = 0.02, respectively). Presence of clonal heterogeneity in driver mutations was not prognostic for OS (P = 0.71). Patients who were found to have 4 or more driver mutations were associated with significantly worse OS (P = 0.014). None of the patients with 4 or more driver mutations had SF3B1 mutation. Multivariate Cox proportional hazard regression analysis considering dichotomized variables relevant to IPSS-R classification (absolute neutrophil count < 0.8 x 103 / µ l, hemoglobin < 8 g/dL, platelet count < 50 x 103 / µ l, and bone marrow blast > 10%, and poor or very poor risk cytogenetics), SF3B1 mutation, DNMT3A mutation, TP53 mutation, and the number of driver mutations (≥ 4) revealed that the presence of 4 or more driver mutations (HR = 2.72 95% CI: 1.34-5.53, P = 0.06), platelet count < 50 x 103 / µ l (HR = 4.73, 95% CI: 2.53-8.85, P < 0.001), and TP53 mutation (HR = 3.34, 95% CI: 1.65-6.75, P = 0.001) significantly predicted worse OS. Conclusion: With the modern sequencing technology, approximately 90% of MDS patients were found to have at least one known myeloid driver mutation. Presence of 4 or more driver mutations in MDS patients predicted poor response to HMA therapy. Multivariate model incorporating mutation profile showed that the presence of 4 or more driver mutations and TP53 mutation status were significantly prognostic in MDS independent of IPSS-R variables. Screening for driver mutations in MDS has clinical impact and mutation profiles should be incorporated into the existing prognostic model. Disclosures Daver: ImmunoGen: Other: clinical trial, Research Funding. DiNardo:Novartis: Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 609-609
Author(s):  
Daniel Noerenberg* ◽  
Larry Mansouri* ◽  
Emma Young ◽  
Frick Mareike ◽  
Maysaa Abdulla ◽  
...  

Abstract Deregulated NF-κB signaling is a hallmark of most, if not all, lymphoid malignancies, and recurrent gene mutations in both the canonical and non-canonical NF-κB pathway are known to lead to NF-κB activation. However, the full compendium of NF-κB gene mutations in lymphoid malignancies remains to be elucidated. Recently, we reported a 4-bp truncating mutation in the NFKBIE gene, which encodes IκBε, a negative regulator of NF-κB, in patients with chronic lymphocytic leukemia (CLL). The NFKBIE deletion was enriched in clinically aggressive CLL patients (7-8%) and associated with a worse clinical outcome. At the functional level, NFKBIE-deleted CLL showed reduced IκBε levels and decreased p65 inhibition, along with increased phosphorylation and nuclear translocation of p65, compared to wildtype patients. Preliminary data has indicated an increased frequency of NFKBIE aberrations in other lymphoid malignancies as well. To explore this further, we screened for NFKBIE deletions in a large cohort of patients diagnosed with a range of different lymphoid neoplasms. Overall, NFKBIE deletions were identified in 76 of 1414 patients (5.4%). While NFKBIE deletions were relatively infrequent in patients diagnosed with follicular lymphoma (3/225, 1.3%), splenic marginal zone lymphoma (3/175, 1.7%), and T-cell acute lymphoblastic leukemia (1/94, 1.1%), moderate frequencies were observed among diffuse large B-cell lymphoma (18/521, 3.5%), mantle cell lymphoma (8/189, 4.2%), and primary CNS lymphoma (1/34, 2.9%) patients. In contrast, a remarkably high frequency of NFKBIE deletions (41/176 cases, 23%) was observed among primary mediastinal B-cell lymphoma (PMBL) patients. Noteworthy, the prevalence of NFKBIE-deleted PMBL cases was similar in the different contributing centers. All PMBL patients in the present series received a CHOP based treatment regime; in ~75% of cases rituximab was added and ~25% were treated with dose intensified schemes. For the latter, the vast majority of patients received CHOEP, while individual cases were treated with MegaCHOEP, DA-EPOCH or ACVBP. Regarding clinicobiological associations, there were no significant differences between NFKBIE-deleted and wildtype PMBL patients with respect to age, sex, Ann Arbor stage, IPI risk-groups, extranodal or bone marrow involvement, bulky disease, and LDH elevation. However, NFKBIE-deleted patients were more likely to be refractory to primary chemotherapy (31% vs. 3%, P=.001) and had a shorter overall survival compared to wildtype patients (5-year overall survival: 63% vs 84%, P=.013). In multivariate analysis (including age, gender, Ann Arbor stage, IPI, and NFKBIE mutation status), NFKBIE mutation status (95% CI: 1.23-10.61; HR: 3.61; P=0.020) remained an independent factor for poor prognosis. In summary, we document NFKBIE deletions as a common genetic event across B-cell malignancies, albeit at varying frequencies. The high frequency of NFKBIE deletions in PMBL alludes to the critical role of this aberration in the pathophysiology of the disease. NFKBIE deletions were associated witha worse clinical outcome, hence potentially representing a novel poor-prognostic marker in PMBL. *Contributed equally as first authors. **Contributed equally as senior authors. Disclosures Stamatopoulos: Gilead: Consultancy, Honoraria, Research Funding; Abbvie: Honoraria, Other: Travel expenses; Novartis: Honoraria, Research Funding; Janssen: Honoraria, Other: Travel expenses, Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4668-4668 ◽  
Author(s):  
Richard J Lin ◽  
Caleb Ho ◽  
Patrick Hilden ◽  
Juliet Barker ◽  
Sergio Giralt ◽  
...  

Abstract TP53 alterations detected by chromosome 17p deletion, oncogenic mutation, or p53 protein overexpression portend extremely poor prognosis for patients with mantle cell lymphoma (MCL), despite aggressive upfront therapy including consolidative high-dose therapy autologous stem cell transplant and; more recently, novel agents such as ibrutinib or lenalidomide. We reviewed outcomes of 42 patients with available TP53 status who had received reduced-intensity/non-myeloablative conditioning (RIC/NMA) allogeneic hematopoietic cell transplant (allo-HCT) for MCL at our institution and examined the impact of TP53 alteration and other clinical factors on outcomes. Nineteen patients had TP53 alterations including 17p deletion (n=3), oncogenic mutation (n=7), and strong p53 protein overexpression by immunohistochemistry (n=9). There were no differences in demographic or clinical characteristics among patients with and without TP53 alterations (Table 1). With a median follow-up for survivors of 23 months (range 4-119), the one-year overall survival (OS) and progression-free survival (PFS) is 87% (95% CI 72-94) and 74% (95% CI 57-85), respectively (Figure 1A). The one-year cumulative incidence of relapse and non-relapse mortality is 16% (95% CI 6-29) and 10% (95% CI 3-22), respectively (Figure 1B). The cumulative incidences of grade II-IV acute GVHD is 38% at 180 days (95% CI 24-53), and of any chronic GVHD is 33% at three-year (95% CI 17-49). Importantly, there is no statistically significant difference in OS (p=0.582, Figure 1C), and cumulative incidence of relapse (p=0.715, Figure 1D) among patients with and without TP53 alterations. In univariate and multivariate analyses, we find that only Ki67 >30% is a significant predictor of PFS (HR 4.1, 95% CI 1.1-15.5, p=0.024), and that only Karnofsky Performance Status (KPS) <90 is a significant predictor of OS (HR 4.9, 95% CI 1.4-17, p=0.01). This is the first report that RIC/NMA allo-HCT could overcome the negative prognostic impact of TP53 alterations in MCL with relatively low incidence of transplant-related mortality. While requiring prospective evaluation, our data supports the consideration of RIC/NMA allo-HCT for MCL patients with TP53 alterations. Disclosures Ho: Invivoscribe, Inc.: Honoraria. Hamlin:Portola: Consultancy. Perales:Novartis: Other: Personal fees; Incyte: Membership on an entity's Board of Directors or advisory committees, Other: Personal fees and Clinical trial support; Merck: Other: Personal fees; Takeda: Other: Personal fees; Abbvie: Other: Personal fees. Sauter:Juno Therapeutics: Consultancy, Research Funding; Sanofi-Genzyme: Consultancy, Research Funding; Spectrum Pharmaceuticals: Consultancy; Novartis: Consultancy; Precision Biosciences: Consultancy; Kite: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document