Abstract
While there is clear evidence to suggest poorer outcome associated with multi-hit (MH) TP53 mutation (TP53MT) compared to a single-hit (SH) mutation in lower-risk myelodysplastic syndrome (MDS), data are conflicting in both higher-risk MDS and acute myeloid leukemia (AML). We conducted an in-depth analysis utilizing data from ten US academic institutions to study differences in molecular characteristics and outcomes of SH (N=139) versus MH (N=243) TP53MT AML. Complex cytogenetics were more common in MH than in SH TP53MT AML (P<0.001); whereas ASXL1 (P<0.001), RAS (P<0.001), splicing factor (P=0.003), IDH1/2 (P=0.001), FLT3 ITD (P<0.001) and NPM1 (P=0.005) mutations clustered significantly with SH TP53MT AML. Survival after excluding patients who received best supportive care alone was dismal but not significantly different between patients with SH or MH disease (event-free survival: 3.0 vs. 2.20 months, respectively, P=0.22; overall survival: 8.50 vs. 7.53 months, respectively, P=0.13). In multivariable analysis, IDH1 mutation and allogeneic hematopoietic stem cell transplantation as a time-dependent covariate were associated with superior event-free survival (hazard ratio [HR]=0.44, 95% confidence interval [95% CI]: 0.19-1.01, P=0.05 and HR=0.34, 95% CI: 0.18-0.62, P<0.001) and overall survival (HR=0.24, 95% CI: 0.08-0.71, P=0.01 and HR=0.28, 95% CI: 0.16-0.47, P<0.001). Complex cytogenetics (HR=1.56, 95% CI: 1.01-2.40, P=0.04) retained an unfavorable significance for overall survival. Our analysis suggests that MH TP53MT is less relevant in independently predicting outcomes in patients with AML than in those with MDS.
Introduction
TP53 is the most frequently mutated gene across all malignancies and is associated with a poor prognosis across many cancer types with suboptimal responses to standard-of-care therapies.1,2 TP53-mutated (TP53MT) acute myeloid leukemia (AML) is strongly associated with large structural and complex cytogenetic abnormalities, often seen among recipients of prior cytotoxic therapies.3-8 Despite the increasing availability of novel therapies, the median overall survival (OS) of patients with TP53MT AML remains in the range of 6-9 months, irrespective of therapy intensity.9-13 Single-hit TP53MT is associated with clonal hematopoiesis and may not be directly leukemogenic unless accompanied by subsequent hits that could be secondary to cytotoxic stress.4,14,15 There are conflicting reports regarding the prognostic impact of allelic state, specifically bi-allelic alteration/“multi-hit” (MH) TP53MT versus mono-allelic/ “single-hit” (SH) TP53MT among patients with myeloid neoplasms.10,16-18 Bernard et al. performed extended genetic profiling in a large cohort of patients with myelodysplastic syndrome (MDS) and showed that not all TP53MT have equivalent impact on survival.19 Patients with MDS harboring SH TP53MT had similar outcomes to their counterparts with TP53 wild-type disease. Conversely, multiple hits caused by either multiple mutations of TP53/copy-neutral loss of heterozygosity or mono-allelic TP53MT with deletion of the other TP53 allele were associated with inferior clinical outcomes. However, in patients with high-risk MDS with excess blasts or AML, it was recently demonstrated that TP53 allelic state (SH vs. MH TP53MT) did not predict differences in clinical outcome.10 The authors concluded that further risk stratification by TP53 allelic state may be less relevant among patients with advanced MDS or AML. Here, we present real world data on a large cohort of patients with TP53MT AML and report their clinical characteristics, therapy received, and outcome based on TP53 allelic state.
Methods
We conducted a retrospective study through the Consortium on Myeloid Malignancies and Neoplastic Diseases (COMMAND) consortium (a collaboration of acute leukemia experts from 10 US academic institutions) to analyze the prognostic impact of MH versus SH TP53MT on outcomes of adult (≥18 years) patients with AML. A total of 382 adults with TP53MT (139 SH, 243 MH) AML who were diagnosed between November 2012 and May 2023 were evaluated, and their baseline characteristics, molecular profile, and treatment outcomes were compared based on SH versus MH TP53MT status. The current cohort of 382 patients was increased from the 291 patients who were included in our previous publication;9 furthermore, the current cohort has more robust TP53 gene annotation data and longer follow-up. These features of the cohort reported here enable a more comprehensive evaluation regarding the impact of TP53 mutation burden on clinical outcome.
MH TP53MT was defined by the presence of two or more distinct TP53MT regardless of variant allele frequency (VAF) or a single TP53MT associated with (i) cytogenetic abnormalities involving chromosome 17p (e.g. abnormality of 17p or monosomy 17) or (ii) a VAF of ≥55%, as previously reported by Grob et al.10 Loss of heterozygosity was not assessed in all patients in this dataset.
AML was diagnosed as per the 2016 World Health Organization classification.20 Response to treatment was defined according to 2017 European LeukemiaNet consensus guidelines.21 Next-generation sequencing was performed at diagnosis using DNA extracted from bone marrow aspirate specimens with post-sequencing analysis of tumor-associated mutations. Next-generation sequencing testing was developed, and its performance characteristics determined by the participating institutions in compliance with Clinical Laboratory Improvement Amendments requirements. The next-generation sequencing panel had a sensitivity of ≥5% VAF with a minimum depth coverage of 250x.
The study was conducted after obtaining approval from the Institutional Review Board, adhering to the ethical standards of the Declaration of Helsinki of 1975, as revised in 2000.
Statistical analysis
Continuous variables are summarized as the median (range) while categorical variables are reported as frequency (percentage). Duration of response (complete [CR] or complete with incomplete blood count recovery [CRi]) was defined from the time of onset of response to progression or death due to any reason, whichever occurred earlier. The Kaplan-Meier method was used to estimate event-free survival (EFS), defined as time from diagnosis to relapse or death. The median overall survival (OS) was calculated from time of diagnosis to death or last follow-up. Cox proportional hazards regression models were used to determine the univariate and multivariate predictors of overall mortality and progression. Allogeneic hematopoietic stem cell transplantation (HSCT) was treated as a time-dependent covariate. Multivariable models included all significant univariate predictors. All tests were two-sided with a P value <0.05 considered statistically significant.
Results
Baseline characteristics
A total of 382 adult patients with TP53MT AML (SH, 139; MH, 243) were identified. Among the 243 patients with MH TP53MT, 57 patients had multiple TP53MT, 58 patients had TP53MT with VAF of ≥55%, and 128 patients had single TP53MT associated with cytogenetic abnormalities involving chromosome 17p (e.g., abnormality of 17p or monosomy 17). The median age was 67 (range, 23-90) and 66.5 (range, 18-97) years in the SH and MH TP53MT AML groups, respectively (P=0.86) (Table 1). Thirty-nine (33%) and 70 (29%) patients had secondary AML in the SH and MH groups, respectively (P=0.34). Among these 109 patients with secondary AML, 11 (10%) patients had JAK2-mutated myeloproliferative neoplasm in blast phase, of whom four (3%) were in the SH group and seven (3%) in the MH group (P=0.82). The median TP53MT VAF was 22% (range, 5-49%) and 50% (range, 5-98%) in the SH and MH TP53MT AML groups (P<0.001), respectively. The proportion of patients with complex cytogenetics was higher in the MH group than in the SH group (93% vs. 58%, P<0.001). In subgroup analysis, we looked at baseline characteristics of patients with IDH1 or IDH2 co-mutated AML. Patients with secondary AML had a higher proportion of IDH2-mutated disease than IDH1-mutated disease (46% vs. 27%, P=0.15) and complex cytogenetics (54% vs. 27%, P=0.24), but these differences were not statistically significant (Online Supplementary Table S1).
Molecular profile and somatic co-mutation pattern
An overview of TP53 domains, distribution of TP53 variants and position on the TP53 protein are illustrated in Figure 1. The occurrences of somatic co-mutations were comparable between the SH (67%) and MH (60%) groups (P=0.22). ASXL1 (16% vs. 7%, P<0.001), RAS (15% vs. 6%, P<0.001), splicing factor (12% vs. 4%, P=0.003), IDH1/2 (11% vs. 4%, P=0.001), FLT3-ITD (11% vs. 2%, P<0.001) and NPM1 (6% vs. 1%, P=0.005) mutations were more frequent in the SH group than in the MH group. The somatic co-mutation patterns and frequency of co-mutations in the SH and MH groups, are illustrated in Figure 2 and Online Supplementary Figure S1, respectively. Eleven (46%) patients had IDH1, and 13 (54%) patients had IDH2 mutations. Two (18%) and seven (53%) patients with IDH1 and IDH2 mutations, respectively, had MH TP53MT. There were no differences in the co-mutational patterns among patients with IDH1/IDH2 co-mutated disease with the lone exception of JAK2 mutations, which were more common in the IDH2 co-mutated group (38.5% vs. 0%, P=0.04) (Online Supplementary Table S1).
Treatment and outcome
A significantly higher proportion of patients in the MH group received hypomethylating agents plus venetoclax compared to the SH group (29% vs. 19%, P=0.01). However, the proportion of patients who received intensive chemotherapy, hypomethylating agent-based therapy or other low-intensity chemotherapy (low-dose cytarabine, IDH2 inhibitor alone or an investigational agent) were comparable between the two groups (Table 1). The response rates (CR/ CRi) were comparable between the SH and MH groups (28% vs. 22%, P=0.21). Among the 91 (26%) patients with CR/CRi, 28 (31%) were did not have measurable residual disease (MRD), assessed by flow cytometry, after induction. The MRD-negative CR rates with intensive versus non-intensive chemotherapy were not significantly different (10% vs. 7%, respectively, P=0.78). Similarly, a comparable proportion of patients underwent allogeneic HSCT after induction (12% vs. 14%, P=0.53). In subgroup analysis, there was a significant difference in response rate between patients with IDH1 co-mutated disease (54.4%) and those with IDH2 co-mutated disease (0%) (P=0.003) (Online Supplementary Table S1). The median duration of response was 7.77 versus 12.83 months in the SH and MH groups, respectively (P=0.73) (Figure 3A).
Predictors of response
Predictors of response (CR/CRi) to induction chemotherapy were evaluated, and the results are summarized in Online Supplementary Table S2. The co-occurrence of RAS (NRAS or KRAS) (P=0.02) and IDH2 mutations (P=0.03) had a negative impact on response rate. Conversely, the co-occurrence of IDH1 mutation (P=0.02) and induction with a hypomethylating agent plus venetoclax (P<0.001) was associated with better responses. In this cohort of adverse-risk TP53MT AML, age ≥65 years (P>0.99), secondary AML (P=0.58), therapy-related AML (P>0.99), and complex cytogenetics (P>0.99) did not have significant impacts on achieving response.
Event-free survival
Considering the significantly higher proportion of patients receiving supportive care alone in the SH group compared to the MH group, we excluded these patients from the survival analysis. The median EFS in months was not significantly different between the SH and MH groups (3.0 and. 2.20, respectively, P=0.22) (Figure 3B). However, there was a statistically significant difference in EFS between the SH and MH groups (3.0 vs. 2.13, P=0.02), utilizing the definition of MH as per the International Consensus Classification, i.e., two distinct TP53MT with VAF >10% or a single TP53MT with (i) 17p deletion; (ii) VAF of >50%; or (iii) copy-neutral loss of heterozygosity at the 17p TP53 locus.22 In univariate analysis for EFS (Online Supplementary Table S3), complex cytogenetics adversely affected outcome (P=0.04). In contrast, ASXL1 mutation (P=0.02), IDH1 mutation (P=0.01), hypomethylating agent plus venetoclax induction (P<0.001), and allogeneic HSCT as a time-dependent covariate (P<0.002) were associated with favorable EFS in univariate analysis. In multivariable analysis for EFS, IDH1 co-mutation (HR=0.44, 95% CI: 0.19-1.01, P=0.05), hypomethylating agent plus venetoclax induction (HR=0.53, 95% CI: 0.41-0.70, P<0.001) and allogeneic HSCT (HR=0.34, 95% CI: 0.18-0.62, P<0.001) retained statistically significant associations with favorable outcomes.
Overall survival
After excluding patients who received supportive care alone, we calculated the median OS. The median OS in months was not significantly different between the SH and MH groups (8.50 vs. 7.53, respectively, P=0.13) (Figure 3C). Likewise, we did not observe a significant difference in OS between the SH and MH groups (8.0 vs. 8.0, respectively, P=0.32), utilizing the MH definition as per the International Consensus Classification. We looked at the impact of complex cytogenetics on OS in the SH and MH groups and found that OS was better in the SH and MH subgroups without complex cytogenetics (9.97 and 10.07 months, respectively) than in those with complex cytogenetics (6.2 and 7.13 months, respectively) (P=0.008) (Figure 3D). We performed landmark analysis from the time of achievement of CR/CRi until last follow-up or death; allogeneic HSCT recipients had better OS compared to non-allogeneic HSCT recipients in both the SH group (not reached vs. 9.63 months, respectively) and the MH group (24.3 vs. 9.6 months, respectively) (P=0.001) (Figure 3E). In another subset analysis among transplanted patients, those who were transplanted in MRD-negative CR (N=12) had a longer median OS compared to those with MRD-positive disease (N=43) (46.1 vs. 25.47 months, respectively, P=0.15) although the difference was not statistically significant probably due to a smaller sample size. We performed a similar analysis to look at OS in relation to complex cytogenetics and TP53 allelic state among patients who achieved CR/CRi and did or did not receive allogeneic HSCT. In subset analysis, we looked at the impact of co-occurring complex cytogenetics on survival outcome of patients in the SH and MH groups. The median OS was 23.6 months, not reached, 20.2 months and not reached in patients with SH and complex cytogenetics, SH without complex cytogenetics, MH with complex cytogenetics and MH without complex cytogenetics, respectively (P=0.18) (Figure 3F). Between patients with SH TP53MT, those who received intensive chemotherapy induction had significantly better outcome compared to those who received non-intensive chemotherapy with a median OS of 9.97 versus 5.82 months, respectively (P=0.04). However, the benefit of intensive chemotherapy compared to non-intensive chemotherapy in improving OS in MH TP53MT was less clear with a median OS of 8.03 versus 6.7 months, respectively (P=0.07).
In univariate analysis for OS (Online Supplementary Table S4), age as a continuous variable (every 10 years) (P=0.02), complex cytogenetics (P=0.002), and other low-intensity chemotherapy (P=0.01) were associated with inferior outcomes. RUNX1 mutation (P=0.01), IDH1 mutation (P<0.001), FLT3 ITD mutation (P=0.003), NPM1 mutation (P=0.02), intensive induction (P=0.007) and allogeneic HSCT as a time-dependent co-variate (P<0.001) were associated with favorable OS in univariate analysis. In multivariable analysis for OS, complex cytogenetics (HR=1.56, 95% CI: 1.01-2.40, P=0.04) retained an unfavorable significance, whereas IDH1 mutation (HR=0.24, 95% CI: 0.08-0.71, P=0.01) and allogeneic HSCT (HR=0.28, 95% CI: 0.16-0.47, P<0.001) retained favorable significance.
Discussion
In our real-world, multicenter analysis of a large cohort of patients with TP53MT AML, we did not observe significant differences in remission rates or survival based on TP53 allelic state. We found that distinct myeloid co-mutation patterns exist between patients with SH and those with MH TP53MT AML, with IDH1 co-mutations imparting a favorable prognostic significance, and that the use of allogeneic HSCT associating with improved OS, irrespective of SH or MH TP53MT status.
Recent studies have explored the clinical significance of TP53MT allelic status in patients with MDS and AML.4,10,23 While patients with MDS harboring SH TP53MT tend to have similar outcomes compared to their TP53 wild-type counterparts and better outcomes than those with MH TP53MT, patients with MDS with excess blasts/AML harboring SH or MH TP53MT had comparable outcomes. Similarly to Grob et al.,10 we did not observe significant differences in response rate or survival between patients with SH or MH TP53MT AML. These data suggest that TP53 allelic state in advanced MDS or AML is less relevant in predicting clinical outcome. Similar to what has been observed in MDS studies, patients with SH TP53MT AML had an abundance of somatic co-mutations, while MH TP53MT AML was significantly associated with occurrence of complex cytogenetics.23
IDH1/2 mutations are observed in approximately 20% of patients with AML (IDH1, 6-16%; IDH2, 8-19%).24 IDH1/2 mutations are more frequently seen in elderly AML patients, especially those with diploid or intermediate-risk cytogenetics, and frequently co-occur with FLT3 ITD and NPM1 mutations.25 With the development of venetoclax and IDH1/2 inhibitors, the outcomes of IDH1/2-mutated AML patients have improved significantly, especially those who are ineligible for intensive therapies.26 Interestingly, we observed significantly improved EFS and OS among patients with IDH1 co-mutations and the favorable significance was retained in multivariate analysis. Moreover, only a small proportion of these patients received venetoclax plus a hypomethylating agent as first-line treatment (2/11 [18%]) or as a salvage therapy (1/11 [9%]) and only 2/11 (18%) patients in this subgroup underwent allogeneic HSCT. None of the patients received an IDH1 inhibitor alone or in combination with chemotherapy upfront. One patient each received a hypomethylating agent/venetoclax plus IDH1 inhibitor and an IDH1 inhibitor alone as a salvage therapy with no response. While these findings are intriguing, they need to be validated in a larger group of patients.
Although allogeneic HSCT is universally considered a potentially curative option for patients with adverse-risk AML, earlier studies showed dismal outcomes for patients with TP53MT AML undergoing allogeneic HSCT.27 Lack of benefit was attributed to inability to achieve complete response and persistence of the pre-transplant TP53MT clone. In our earlier report utilizing data from ten US academic centers, we showed that allogeneic HSCT improved survival of patients with TP53MT AML.13 We have now re-confirmed this finding and shown that it holds true irrespective of TP53 allelic state. The multivariable analysis in this study also demonstrated a significantly better EFS associated with induction with a hypomethylating agent plus venetoclax when compared with other therapies. However, this did not translate into improved OS, suggesting evolution of resistant clones that were not suppressed in the long-term by venetoclax plus hypomethylating agent therapy alone, as previously reported.28 Secondly, in a subset analysis we observed better OS with intensive chemotherapy compared to non-intensive chemotherapy induction in the SH and MH subgroups, probably due to the fact that patients eligible for intensive chemotherapy generally have good performance status/fewer co-morbidities and are more likely candidates for allogeneic HSCT. Furthermore, intensive chemotherapy induction did not retain significance for better survival in multivariate analysis.
We acknowledge some limitations of our analysis including a selection bias inherent to a retrospective analysis and some overlap with our prior work.9 However, our current analysis includes 382 patients followed longitudinally, significantly strengthening our previous cohort of 291 patients, with more robust TP53 gene annotation data, and these patients have longer follow-up. This strengthened cohort enabled a more comprehensive evaluation of the impact of TP53 mutation burden on clinical outcome. Second, cases with apparent mono-allelic TP53MT may have hidden clones with bi-allelic TP53 inactivation which were not detected by widely used sequencing methods. Furthermore, although loss of heterozygosity to determine TP53MT allelic state was not assessed in all patients in this dataset, we defined SH and MH TP53MT based on earlier observations by Grob et al.10 Moreover, we did not observe significant differences in survival outcomes using the definition of MH TP53 as per the International Consensus Classification or by Grob et al.10
In conclusion, unlike in lower-risk MDS, we did not find a significant difference in response rate or survival outcomes between patients with SH or MH TP53MT AML, which is consistent with recent reports.10,18 Prospective studies are needed to better understand the effect of TP53 allelic state on the outcomes of patients with TP53MT AML.
Footnotes
- Received January 6, 2024
- Accepted May 21, 2024
Correspondence
Disclosures
TB has served on advisory boards for Pfizer, Morphosys and Takeda. AP has provided consultancy services for AbbVie; has received research funding from Kronos Bio, Pfizer, Celgene/ BMS, and Servier; has received honoraria from AbbVie and BMS and has received institutional research funding from Pfizer and Kronos Bio. VK has served on advisory boards for Novartis and Pfizer. AMZ is a Leukemia and Lymphoma Society Scholar in Clinical Research; has received institutional research funding from Celgene/BMS, AbbVie, Astex, Pfizer, Medimmune/AstraZeneca, Boehringer-Ingelheim, Cardiff Oncology, Incyte, Takeda, Novartis, Shattuck Labs, Geron, and Aprea; has participated in advisory boards, and/ or had a consultancy role with and received honoraria from AbbVie, Pfizer, Celgene/BMS, Jazz, Incyte, Agios, Servier, Boehringer-Ingelheim, Novartis, Astellas, Daiichi Sankyo, Geron, Taiho, Seattle Genetics, BeyondSpring, Takeda, Ionis, Amgen, Janssen, Genentech, Epizyme, Syndax, Gilead, Kura, Chiesi, ALX Oncology, BioCryst, Notable, Orum, Mendus, Foran, Syros, and Tyme; has served on clinical trial committees for Novartis, AbbVie, Gilead, Syros, BioCryst, Abbvie, ALX Oncology, Geron and Celgene/BMS; and has received travel support for meetings from Pfizer, Novartis, and Cardiff Oncology. The other authors have no conflicts of interest to disclose.
References
- Daver NG, Maiti A, Kadia TM. TP53-mutated myelodysplastic syndrome and acute myeloid leukemia: biology, current therapy, and future directions. Cancer Discov. 2022; 12(11):2516-2529. Google Scholar
- Sabapathy K, Lane DP. Therapeutic targeting of p53: all mutants are equal, but some mutants are more equal than others. Nat Rev Clin Oncol. 2018; 15(1):13-30. Google Scholar
- Badar T, Szabo A, Sallman D. Interrogation of molecular profiles can help in differentiating between MDS and AML with MDS-related changes. Leuk Lymphoma. 2020; 61(6):1418-1427. Google Scholar
- Hiwase D, Hahn C, Tran ENH. TP53 mutation in therapyrelated myeloid neoplasm defines a distinct molecular subtype. Blood. 2023; 141(9):1087-1091. Google Scholar
- Haase D, Stevenson KE, Neuberg D. TP53 mutation status divides myelodysplastic syndromes with complex karyotypes into distinct prognostic subgroups. Leukemia. 2019; 33(7):1747-1758. Google Scholar
- Kandoth C, McLellan MD, Vandin F. Mutational landscape and significance across 12 major cancer types. Nature. 2013; 502(7471):333-339. Google Scholar
- Weinberg OK, Siddon A, Madanat YF. TP53 mutation defines a unique subgroup within complex karyotype de novo and therapy-related MDS/AML. Blood Adv. 2022; 6(9):2847-2853. Google Scholar
- Sallman DA, Komrokji R, Vaupel C. Impact of TP53 mutation variant allele frequency on phenotype and outcomes in myelodysplastic syndromes. Leukemia. 2016; 30(3):666-673. Google Scholar
- Badar T, Atallah E, Shallis RM. Outcomes of TP53-mutated AML with evolving frontline therapies: impact of allogeneic stem cell transplantation on survival. Am J Hematol. 2022; 97(7):E232-E235. Google Scholar
- Grob T, Al Hinai ASA, Sanders MA. Molecular characterization of mutant TP53 acute myeloid leukemia and high-risk myelodysplastic syndrome. Blood. 2022; 139(15):2347-2354. Google Scholar
- Rücker FG, Schlenk RF, Bullinger L. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012; 119(9):2114-2121. Google Scholar
- Papaemmanuil E, Gerstung M, Bullinger L. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016; 374(23):2209-2221. Google Scholar
- Badar T, Atallah E, Shallis R. Survival of TP53-mutated acute myeloid leukemia patients receiving allogeneic stem cell transplantation after first induction or salvage therapy: results from the Consortium on Myeloid Malignancies and Neoplastic Diseases (COMMAND). Leukemia. 2023; 37(4):799-806. Google Scholar
- Boettcher S, Miller PG, Sharma R. A dominant-negative effect drives selection of TP53 missense mutations in myeloid malignancies. Science. 2019; 365(6453):599-604. Google Scholar
- Wong TN, Ramsingh G, Young AL. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature. 2015; 518(7540):552-555. Google Scholar
- Zeidan AM, Bewersdorf JP, Hasle V. Prognostic implications of mono-hit and multi-hit TP53 alterations in patients with acute myeloid leukemia and higher risk myelodysplastic syndromes treated with azacitidine-based therapy. Leukemia. 2023; 37(1):240-243. Google Scholar
- Bahaj W, Kewan T, Gurnari C. Novel scheme for defining the clinical implications of TP53 mutations in myeloid neoplasia. Res Sq. 2023. Google Scholar
- Stengel A, Meggendorfer M, Walter W. Interplay of TP53 allelic state, blast count, and complex karyotype on survival of patients with AML and MDS. Blood Adv. 2023; 7(18):5540-5548. Google Scholar
- Bernard E, Nannya Y, Hasserjian RP. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat Med. 2020; 26(10):1549-1556. Google Scholar
- Arber DA, Orazi A, Hasserjian R. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016; 127(20):2391-2405. Google Scholar
- Döhner H, Estey E, Grimwade D. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017; 129(4):424-447. Google Scholar
- Arber DA, Orazi A, Hasserjian RP. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data. Blood. 2022; 140(11):1200-1228. Google Scholar
- Bernard E, Nannya Y, Hasserjian RP. Implications of TP53 allelic state for genome stability, clinical presentation and outcomes in myelodysplastic syndromes. Nat Med. 2020; 26(10):1549-1556. Google Scholar
- DiNardo CD, Ravandi F, Agresta S. Characteristics, clinical outcome, and prognostic significance of IDH mutations in AML. Am J Hematol. 2015; 90(8):732-736. Google Scholar
- Paschka P, Schlenk RF, Gaidzik VI. IDH1 and IDH2 mutations are frequent genetic alterations in acute myeloid leukemia and confer adverse prognosis in cytogenetically normal acute myeloid leukemia with NPM1 mutation without FLT3 internal tandem duplication. J Clin Oncol. 2010; 28(22):3636-3643. Google Scholar
- Pollyea DA, DiNardo CD, Arellano ML. Impact of venetoclax and azacitidine in treatment-naïve patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 2022; 28(13):2753-2761. Google Scholar
- Mohr B, Schetelig J, Schäfer-Eckart K. Impact of allogeneic haematopoietic stem cell transplantation in patients with abnl(17p) acute myeloid leukaemia. Br J Haematol. 2013; 161(2):237-244. Google Scholar
- DiNardo CD, Tiong IS, Quaglieri A. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood. 2020; 135(11):791-803. Google Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.