Original Study
TP53 and IDH2 Somatic Mutations Are Associated With Inferior Overall Survival After Allogeneic Hematopoietic Cell Transplantation for Myelodysplastic Syndrome

https://doi.org/10.1016/j.clml.2017.06.003Get rights and content

Abstract

Background

Next-generation sequencing has identified somatic mutations that are prognostic of cancer.

Patients and Methods

We evaluated the incidence and prognostic significance of somatic mutations in 89 myelodysplastic syndrome (MDS) patients who received an allogeneic hematopoietic cell transplantation. Next-generation sequencing was performed on paraffin embedded bone marrow, which was obtained at a median of 31 days before initiating the preparative regimen.

Results

The 3 most common subtypes of MDS were refractory anemia with excess blasts (RAEB)-1 (35%), RAEB-2 (29%), and refractory cytopenia with multilineage dysplasia (18%). Most patients (91%) received a myeloablative regimen of fludarabine with intravenous busulfan. Somatic mutations (> 0) were identified in 39 (44%) of analyzed samples. The 6 most commonly identified gene mutations were ASXL1 (8%), DNMT3A (8%), RUNX1 (7%), KRAS (6%), IDH2 (4%), and TP53 (4%). The low incidence of mutations in our study sample might be explained by tissue source and stringent variant-calling methodology. Moreover, we speculate that the low incidence of mutations might, perhaps, also be explained by previous azacitidine treatment in 82% of cases. Multivariate analysis identified TP53 (hazard ratio [HR], 3.82; 95% confidence interval [CI], 1.12-13.09; P = .03) and IDH2 mutations (HR, 4.74; 95% CI, 1.33-16.91; P = .02) as predictors of inferior 3-year overall survival.

Conclusion

This study furthers implementation of clinical genomics in MDS and identifies TP53 and IDH2 as targets for pre- or post-transplant therapy.

Introduction

Despite growing understanding of the genetic and molecular basis of myelodysplastic syndrome (MDS) as well as availability of epigenetic therapy, the disease remains incurable unless eligible patients are offered an allogeneic hematopoietic cell transplantation (allo-HCT).1, 2, 3, 4, 5, 6 Nevertheless, only approximately half of the patients who undergo the procedure achieve durable remissions.5, 6 Significant morbidity and mortality occurs despite efforts to optimize patient selection using disease-specific hematopoietic cell transplantation comorbidity indices.7, 8

No randomized data exist for comparisons of allo-HCT versus nontransplant therapies in patients with MDS. Presently, the decision to offer an allo-HCT is on the basis of published decision models that have shown a significant benefit favoring an allo-HCT earlier in the disease course, for patients with intermediate- or higher-risk MDS whether allografted from histocompatible siblings or unrelated donors.9, 10 Published retrospective single-institution or registry studies have also supported the benefit of an allo-HCT to eligible patients with MDS.11, 12, 13, 14 Various clinical models have been developed aimed at improving outcomes in patients who undergo an allo-HCT for MDS by identifying patient's comorbidities7, 15 and clinical characteristics of the disease.16 These models, however, are unable to predict outcomes in individual cases.

Availability of next-generation sequencing (NGS) has helped identify somatic mutations capable of prognosticating outcomes of patients with MDS and acute myeloid leukemia (AML).1, 17, 18 Bejar et al evaluated, retrospectively, 87 subjects who underwent an allo-HCT for MDS, median age 58 (range, 19-73) years, and identified the presence of at least 1 somatic mutations in 80 of 87 (92%) of cases.19 In their series, mutated ASXL1 was the most frequently observed mutation (n = 25/87, 29%).19 Moreover, mutations in TP53, TET2, and DNMT3A were associated with an inferior overall survival (OS) after allo-HCT; and presence of mutated TP53 was described as the most significant predictor of mortality after transplantation with a short median survival of 4.6 months.19 This study highlights the importance of identifying strong predictors of adverse outcomes in patients with MDS who undergo allo-HCT to develop therapeutic strategies aimed at improving outcomes.19 Integrating NGS as a prognostic tool represents a step in the right direction aimed at moving MDS therapeutics to a more personalized approach.

Section snippets

Patient and Treatment Characteristics

Patients were considered eligible for inclusion in this study if they were ≥ 18 years of age, received an allo-HCT for MDS between January 2005 and June 2012, and had available formalin-fixed paraffin embedded (FFPE) bone marrow (BM) samples from the pretransplant workup, which was performed within approximately 30 days from initiation of the preparative regimen. All samples were collected according to a protocol approved by the institutional review board of the University of South Florida.

Patient, Disease, and Transplant-Related Characteristics

Initially, we identified 139 patients who received an allo-HCT during the specified time period. However, 38 were excluded for inability to isolate DNA because of poor quality of FFPE BM samples and 12 because of diagnoses other than MDS (AML = 4, chronic myelomonocytic leukemia = 8). Accordingly, 89 patients (male = 52%), median age 58 (range, 22-74) years, were included in this cohort. The 3 most common MDS types at the time of diagnosis, defined according to the 2008 World Health

Discussion

The presence of mutated TP53 is associated with an inferior OS and a strong trend for worse PFS in patients with MDS who undergo an allo-HCT (Table 2). In our study, the median survival of patients harboring TP53 mutations was only 6.1 months and the 3-year PFS and OS were 0. Lindsley et al showed that TP53 mutations are associated with shorter survival and a shorter time to relapse.22 These findings are comparable with a previous single-institution study that reported a median survival of 4.6

Conclusion

Rapidly expanding knowledge pertaining to the prognostic significance of various gene mutations has only begun to augment disease risk prognostication for MDS. This study furthers the implementation of clinical genomics in this disease and will help incorporate novel therapies, for example IDH2 inhibitors or others, in the post-transplant setting aimed at reducing the risk associated with these mutations.

Disclosure

The authors have stated that they have no conflicts of interest.

Acknowledgments

The authors acknowledge Moffitt tissue core and genomics core and Core Grant P30-CA076292. This work was supported by a grant from Moffitt Cancer Center Foundation to M.A.K-D.

References (27)

  • R. Bejar et al.

    Clinical effect of point mutations in myelodysplastic syndromes

    N Engl J Med

    (2011)
  • C. Cutler

    Timing of allogeneic stem cell transplantation for myelodysplastic syndromes and aplastic anemia

    Hematology Am Soc Hematol Educ Program

    (2014)
  • H.J. Deeg

    Hematopoietic cell transplantation for myelodysplastic syndrome

    Am Soc Clin Oncol Educ Book

    (2015)
  • Cited by (18)

    • Clinical Characteristics, Prognosis, and Treatment Strategies of TP53 Mutations in Myelodysplastic Syndromes

      2022, Clinical Lymphoma, Myeloma and Leukemia
      Citation Excerpt :

      As is shown in Figure 1, the initial literature search included 634 articles. After excluding 62 duplicates and other 552 pieces for the reasons of reviews, case reports, animals or cells experiment, and insufficient data, we finally enrolled 20 primary studies compromising 5067 patients in this Analysis.17,18,8,19-23,9,24-33,34 The sample sizes ranged from 39 to 1514, while the frequency of mutations in TP53 ranged from 5.3% to 74.6%.

    • Myeloablative Conditioning for Allogeneic Transplantation Results in Superior Disease-Free Survival for Acute Myelogenous Leukemia and Myelodysplastic Syndromes with Low/Intermediate but not High Disease Risk Index: A Center for International Blood and Marrow Transplant Research Study

      2020, Biology of Blood and Marrow Transplantation
      Citation Excerpt :

      Owing to an inherent limitation in all retrospective studies, our analysis could not adjust for the clinical decision making factors (eg, specific comorbidities, performance score, disease status, genetic risk) that prompted treating physicians to choose MAC or RIC for each individual study patient. We were unable to adjust for factors such as molecular abnormalities or MRD status that can also affect the risk of relapse and subsequent survival after HCT [5,6,29-36]. However, because no widely used standardized methods for the high-sensitivity quantification of residual disease burden before HCT are yet widely available for patients with AML [5], future studies could prospectively reexamine the role of conditioning intensity on HCT outcomes where both MRD status and molecular genetic risk are considered.

    • TP53 mutation in allogeneic hematopoietic cell transplantation for de novo myelodysplastic syndrome

      2018, Leukemia Research
      Citation Excerpt :

      However, 21% of the enrolled patients had therapy-related MDS, which prevented the identification of de novo MDS-specific genetic mutations [16]. The other study failed to identify any genetic mutations associated with transplant outcomes, but the sample size was relatively small and the inclusion of therapy-related MDS was not clarified [32]. One recent study investigated the outcomes after allogeneic HCT for therapy-related MDS patients and the authors reported that there was no survival difference according to the presence or absence of TP53 mutation [33].

    View all citing articles on Scopus

    This study was presented in part at the 57th annual meeting of the American Society of Hematology on December 7, 2015 (abstract 4382).

    View full text