International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Acute myeloid leukemia (AML) is a highly heterogeneous hematological malignancy that poses significant treatment challenges. Despite considerable advancements in induction and consolidation therapies for AML in recent years, the risk of relapse after achieving complete remission (CR) remains high (1). Specifically, among patients aged <60 years, >50% experienced relapse, whereas relapse rates reached 90% in those aged ≥60 years. This high disease recurrence rate contributes to an aggregated 5-year overall survival (OS) of 30.5% across all cases (2). This underscores the urgent need for effective and safe post-remission treatment strategies to enhance disease-free survival and ultimately OS survival. While consolidation therapy has been established to improve survival outcomes in low-risk patients with AML, maintenance therapy, a standard treatment component for certain hematological malignancies, such as acute lymphoblastic leukemia and acute promyelocytic leukemia (3,4), remains a contentious issue in AML. For patients with AML harboring high-risk factors for recurrence and who have undergone allogeneic hematopoietic stem cell transplantation, maintenance therapy is deemed necessary to mitigate the risk of recurrence (5,6). However, for those with intermediate-to-low-risk disease, maintenance therapy is not yet part of the standard treatment regimen due to insufficient evidence of its benefits. Thus, the exploration of maintenance therapy for patients with intermediate and low-risk AML is of importance.
The combination of azacitidine (AZA) and venetoclax (VEN) has demonstrated synergistic effects, enhancing the anti-leukemic efficacy while potentially reducing drug dosages and associated toxicities (7,8). This VEN and hypomethylating agent regimen has emerged as the standard treatment for elderly or unfit patients with AML who are not suitable for intensive chemotherapy, where unfitness is defined as meeting ≥1 of the following: Eastern Cooperative Oncology Group performance status ≥2; Hematopoietic Cell Transplantation Comorbidity Index ≥3; organ dysfunction; or active infection requiring intravenous antimicrobial therapy (9–11). Although studies have explored the use of AZA or VEN individually for maintenance therapy in AML (12), there is a paucity of research on the application of the combined VEN plus AZA (VEN-AZA) regimen for maintenance therapy in patients with low-to-intermediate-risk AML.
In the present study, a retrospective analysis of 43 patients newly diagnosed with intermediate-to-low-risk AML who underwent maintenance therapy at Beijing Luhe Hospital Affiliated to Capital Medical University (Beijing, China) was conducted to evaluate the efficacy and safety of the combination regimen of VEN-AZA in maintenance therapy for patients with this disease.
A retrospective analysis of patients with primary intermediate-to-low-risk AML admitted to Beijing Luhe Hospital Affiliated to Capital Medical University between March 2018 and June 2024 was conducted. Prognostic risk stratification was conducted according to the 2022 European Leukemia Net AML criteria (13). The inclusion criteria were as follows: i) Patients who remained in CR following induction and consolidation therapy, with blasts of <5% and no evidence of extramedullary tumor infiltration and received either VEN-AZA treatment regimen or no treatment; ii) patients who were not eligible for or unwilling to undergo allogeneic or autologous hematopoietic stem cell transplantation; iii) patients who had not received other targeted drugs; and iv) patients with complete clinical data. The study variables including the patient's age, sex, prognosis stratification, white blood cell (WBC) count, platelet count, lactate dehydrogenase levels, bone marrow blast percentage, chromosomal analysis, fusion genes, gene mutations and minimal residual disease (MRD) monitoring prior to maintenance therapy were retrospectively extracted from electronic medical records.
Diagnostic data collection Bone marrow morphological examination was performed using Wright's staining, with 200 nucleated cells counted under an oil-immersion lens. All molecular analyses, including reverse transcription-quantitative PCR (RT-qPCR) for AML1-ETO, CBFβ-MYH11 and HOX11 expression quantification, as well as next-generation sequencing (NGS)-based mutation profiling of 248 genes, were performed by Hightrust Diagnostics Medical Laboratory (Beijing, China). The RT-qPCR primer sequence information for various genes is shown in Table I. For NGS, libraries were constructed using the Hemaseq™ Myeloid 248 Gene Mutation Detection Kit (Healthy Biotechnology Co., Ltd; cat. no. S0918-S), a hybridization capture-based panel targeting exons and flanking ±20 bp intronic regions of 248 genes, including core driver genes [such as fms related receptor tyrosine kinase 3 (FLT3), nucleophosmin 1 (NPM1) and CCAAT enhancer binding protein α (CEBPA)], epigenetic regulators [(such as DNA methyltransferase 3α (DNMT3A), tet methylcytosine dioxygenase 2 (TET2), isocitrate dehydrogenase [NADP(+)] 1/2 (IDH1/2)], signaling pathway genes (such as Janus kinase 2 and calreticulin) and spliceosome complex genes (such as splicing factor 3b subunit 1 and serine and arginine rich splicing factor 2).
For bone marrow MRD detection, a Canto II flow cytometer (Becton, Dickinson and Company) with Diva software was used to acquire and analyze the data. Antibody reagents from Becton, Dickinson and Company and Beckman Coulter, Inc. were used to label CD34, CD117, CD38, HLA-DR, CD33, CD13, CD15, CD14, CD4, CD7, CD19 and CD56. After collecting 500,000 CD45+ cells, abnormal cell populations were identified by combining leukemia-associated immunophenotype and ‘different from normal’ approaches. The threshold was set at 0.1% as per the European LeukemiaNet recommendations to distinguish negative and positive results (14). Additionally, RT-qPCR targeting AML1-ETO and CBFβ-MYH11 fusion-gene transcripts was used for MRD detection.
Patients in the maintenance therapy group received VEN-AZA maintenance therapy 1 month after consolidation therapy. The specific VEN-AZA dosing regimen was as follows: Subcutaneous injection of 75 mg/m2 AZA for 7 consecutive days and the dose of VEN was reduced from the standard 400 mg daily to 200 mg on days 1–21. If grade 4 myelosuppression (based on the Common Terminology Criteria for Adverse Events version 5.0) occurred during treatment (15), the VEN treatment was paused and supportive treatments such as leukocyte elevation, anti-infection measures or component blood transfusions were provided. One treatment cycle lasted 2–3 months. During the maintenance treatment period, an assessment was conducted per treatment cycle. The methods for detecting MRD included qPCR for detecting fusion genes and flow cytometry to identify leukemia-associated abnormal phenotypes in the bone marrow cells of patients. The primary efficacy endpoint was progression-free survival (PFS), defined as the time from the onset of the disease to recurrence, death or end of follow-up.
Continuous variables are presented as the median and range (minimum to maximum). Intergroup comparisons were conducted using the Kruskal-Wallis rank-sum test. For categorical variables, proportional differences were analyzed via Fisher's exact test when any expected cell count was <10; otherwise, the χ2 test was applied (Table II). The impact of maintenance therapy on PFS was evaluated using Kaplan-Meier survival curves and log-rank tests. Differences in PFS were assessed using univariate and multivariate Cox proportional hazards regression analysis. P<0.05 was considered to indicate a statistically significant difference. Statistical analysis was performed using Empower(R) (www.empowerstats.com; X&Y solutions, Inc.) and R version 3.6.1 (http://www.R-project.org).
The present study retrospectively analyzed 43 cases of newly diagnosed patients with intermediate-to-low-risk AML who remained in CR following the completion of induction and consolidation therapy Among them, there were 10 patients in the maintenance therapy group who received intensive treatment during the induction phase and 11 patients who received low-intensity treatment [based on hypomethylating agents (HMAs) or low-dose cytarabine]. Thus, there were 22 patients who received maintenance therapy. The median number of maintenance treatment cycles was 8 (range, 2–18). Among these patients, 59.09% (n=13) achieved MRD-negative (MRDneg) status following induction and consolidation therapy. There were 21 patients who did not receive maintenance therapy, with 66.67% (n=14) achieving MRDneg status following the induction and consolidation therapy. There was no significant difference in the MRDneg status between the off-maintenance therapy group and the maintenance therapy group at enrollment. The gene mutations with the highest frequency in the 43 patients were NPM1, CEBPA, FLT3, DNMT3A, KIT, ASXL transcriptional regulator 1 (ASXL1), ASXL2, NRAS, IDH1, IDH2 and TET2. The comparison of basic clinical characteristics between the two groups is shown in Table II. The proportion of patients with CEBPA mutations was higher in the off-maintenance therapy group (33.33% vs. 4.55%; P=0.021), and the proportion of patients in the favorable group was also higher in the off-maintenance therapy group (61.90 vs. 31.82%; P=0.048). During the study period, the most common grade 3–4 hematological adverse events in the maintenance treatment group were neutropenia (n=2) and thrombocytopenia (n=4), and the most common non-hematological adverse event was respiratory infection (Table III). Grade 3–4 adverse events were experienced by 9 patients during cycles 1–2 of treatment. No deaths occurred during the maintenance treatment period.
Clinical and pathological parameters predictive of PFS were further investigated through univariate and multivariate Cox regression models. In the univariate analysis, maintenance therapy selection, WBC and age were significantly associated with PFS (Table IV). Patients with AML who did not accept maintenance therapy showed a significantly unfavorable PFS. With a median follow-up of 29.6 months (7–74), the median PFS was not reached in patients with AML receiving maintenance therapy, while the median PFS time for those not receiving maintenance therapy was 47.3 months (P=0.0034; Fig. 1D). The PFS time decreased in patients with WBC ≥50×109/l and those aged ≥60 (P=0.0012 and P=0.0481, respectively; Fig. 1A and B). No difference in PFS was observed between the intermediate-risk and low-risk patients (P=0.9892; Fig. 1C).
The multivariate Cox regression model was employed to examine the association between VEN-AZA maintenance therapy and PFS, with adjustments made for potential confounding factors (Table V). In both the unadjusted and adjusted models, it was observed that patients with AML who underwent VEN-AZA maintenance treatment exhibited a decreased risk of the primary endpoint event, compared with those not receiving maintenance treatment. In the unadjusted model, the hazard ratio (HR) was determined to be 0.09, with a 95% confidence interval (CI) of 0.01–0.69. In the Adjusted I model, the adjustment factors included the age of the patients at the time of diagnosis, which was categorized as either ≥60 years old or <60 years old, and the WBC count was specified as ≥50×109/l or <50×109/l. The corresponding HR was found to be 0.1, with a 95% CI of 0.01–0.85. In the Adjusted II model, the adjustment factors comprised the age of the patients at diagnosis (≥60 years old or <60 years old), the WBC count (≥50×109/l or <50×109/l), prognostic stratification and mutations in the DNMT3A, NRAS, IDH2, IDH1, TET2, ASXL1 and ASXL2 genes, as well as MRD prior to maintenance treatment. The HR was calculated to be 0.06, with a 95% CI of 0.00–0.77.
Table V.Association between VEN-AZA maintenance therapy and the endpoint of acute myeloid leukemia recurrence in multivariate Cox proportional-hazards analysis. |
Maintenance therapy for AML plays a pivotal role in reducing the recurrence risk and enhancing patient survival rates. The VIALE-A study (16) demonstrated that combination treatment with VEN-AZA can significantly improve treatment outcomes. The VEN-AZA regimen is typically designed as a long-term treatment, which persists until disease progression or the emergence of unacceptable toxicity (17). In the present study, patients with intermediate-to-low-risk AML attained CR following consolidation therapy. Among those continuing VEN-AZA maintenance therapy, only 1 case experienced disease progression, with PFS not reached, showing a significantly longer PFS compared with the non-maintenance group (median PFS of 47.3 months). Even after adjusting for age, WBC count, prognostic stratification and prognosis-related gene mutations, the relapse risk of maintenance-treated patients remained significantly lower. Pratz et al (18) demonstrated the importance of MRD in a cohort of patients with a longer follow-up (median >20 months). Those who achieved CR without measurable residual disease also showed a longer duration of remission, OS and event-free survival compared with patients with MRD positivity. The VIALE-A study (16) demonstrated that patients responsive to VEN-AZA who achieved CR with MRD negativity had a median OS time of ~3 years. The study by Chua et al (19) confirmed that patients receiving VEN-based treatment for 12 months and achieving MRD negativity at the end of treatment had the potential for durable treatment-free remission (TFR). Deep remission is a key determinant of long-term survival in patients with AML. During continuous VEN-AZA treatment in the present study, the MRD of 8 patients turned negative, achieving molecular remission, with no relapses. By contrast, 7 patients had persistent positive MRD following consolidation and 6 exhibited relapse. This is consistent with previously reported results that indicated that late-stage MRDneg responses were associated with an improved prognosis (20). This suggested that intermediate- or low-risk patients may benefit from VEN-AZA maintenance, likely due to deep remission induced by continuous treatment, effectively preventing relapse.
The maintenance treatment regimen for AML should effectively prevent recurrence while avoiding severe chemotherapy-related adverse reactions. A previous study reported that the incidence of any-grade infections was 84% in the combination therapy group and 67% in the single-agent AZA group. The incidence of severe adverse events was 83% in the combination therapy group, higher than the 73% in the single-agent AZA group (21). Of note, in the present study, no deaths or severe adverse events occurred during the maintenance treatment of patients. The most common grade 3–4 adverse events were neutropenia, thrombocytopenia and respiratory tract infections. Most adverse reactions occurred in the first two cycles of maintenance treatment, and then the frequency gradually decreased, which may have been associated with the dose reduction of VEN or the adjustment of the administration time. As demonstrated in the retrospective analysis by Xin et al (22), in patients who responded to the combination therapy of HMAs and VEN, the incidence of infectious complications was relatively high, and the infection-related mortality was also at a high level. The infection-related mortality has an adverse impact on the OS of this vulnerable patient population with AML. Elderly patients with AML or those unfit for intensive chemotherapy are more likely to be affected by treatment-related toxicities during long-term treatment (23). Therefore, for patients with intermediate- or low-risk AML who achieve deep (MRD ≤0.01%) and sustained CR, attempting to discontinue treatment and obtaining the benefits of treatment-free survival (TFS) could be a feasible strategy.
However, whether discontinuing treatment is the best strategy for achieving an improvement in quality of life and TFR remains controversial (24). In the present study, the median number of maintenance treatment cycles was 8 (range, 2–18), yet we were unable to determine the minimum number of VEN-AZA treatment cycles associated with a longer TFR. As reported in the study by Garciaz et al (25), patients who had received >10 previous cycles of treatment had the longest TFS, and a minimum of 5 cycles were associated with an acceptable TFR. Therefore, further exploration to determine the optimal number of cycles and dosage for maintenance therapy is needed.
Although the present study demonstrated the advantages of VEN-AZA maintenance therapy in reducing the recurrence risk and prolonging survival in patients with intermediate- or low-risk AML, it should be noted that the study has limitations. The small sample size increases the likelihood of failing to detect true treatment effects. As a retrospective study, a number of confounding factors, including prior consolidation chemotherapy regimens and prophylactic antibiotic choices, were not included, which may have inflated the observed treatment benefits. Additionally, the limited follow-up duration might have led to an underestimation of late toxicities. Future research should focus on conducting large-scale, long-term and strictly controlled clinical trials to accurately determine the minimum number of VEN-AZA maintenance treatment cycles required to achieve durable TFR without compromising treatment efficacy. Additionally, given that existing studies have shown that molecular changes, such as NPM1 and IDH mutations, are associated with a higher benefit from VEN-AZA treatment (26), identifying predictive biomarkers to accurately screen out patients who are most likely to benefit from VEN-AZA maintenance therapy will lay the foundation for developing truly individualized treatment strategies.
Not applicable.
The present study was funded by Youth Support Project of Luhe Hospital (grant no. LHYH2024-LC07).
The data generated in the present study may be requested from the corresponding author.
DZ and XD conceptualized the study, designed the methodology and interpreted data. DZ and HZ conducted the statistical analyses and validated the analytical models. XD drafted the manuscript and coordinated revisions. YZ, XC and JQ completed the acquisition and preliminary interpretation of clinical data. TC participated in the study design. HZ adjudicated clinical endpoints, resolved discrepancies in adverse event attribution and critically revised the manuscript for scientific accuracy. HZ and XC confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.
This study was approved by the Ethics Committee of the Affiliated Beijing Luhe Hospital of Capital Medical University (Beijing, China; approval no. 2024-KY-108). Given the retrospective nature of the study and the use of de-identified patient data, the requirement for informed consent was waived by the Institutional Review Board. The study was conducted in accordance with the ethical standards of the Declaration of Helsinki and its later amendments.
Not applicable.
The authors declare that they have no competing interests.
|
Yuan XL, Lai XY, Wu YB, Yang L, Shi J, Liu L, Zhao Y, Yu J, Huang H and Luo Y: Risk factors and prediction model of early relapse in acute myeloid leukemia after allogeneic hematopoietic cell transplantation. Blood. 140:12915. 2022. View Article : Google Scholar | |
|
DiNardo CD, Erba HP, Freeman SD and Wei AH: Acute myeloid leukaemia. Lancet. 401:2073–2086. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Toksvang LN, Lee SHR, Yang JJ and Schmiegelow K: Maintenance therapy for acute lymphoblastic leukemia: Basic science and clinical translations. Leukemia. 36:1749–1758. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sanz MA, Fenaux P, Tallman MS, Estey EH, Löwenberg B, Naoe T, Lengfelder E, Döhner H, Burnett AK, Chen SJ, et al: Management of acute promyelocytic leukemia: Updated recommendations from an expert panel of the European LeukemiaNet. Blood. 133:1630–1643. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Kinsella FAM, Maroto MAL, Loke J and Craddock C: Strategies to reduce relapse risk in patients undergoing allogeneic stem cell transplantation for acute myeloid leukaemia. Br J Haematol. 204:2173–2183. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Molica M, Breccia M, Foa R, Jabbour E and Kadia TM: Maintenance therapy in AML: The past, the present and the future. Am J Hematol. 94:1254–1265. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Tsao T, Shi Y, Kornblau S, Lu H, Konoplev S, Antony A, Ruvolo V, Qiu YH, Zhang N, Coombes KR, et al: Concomitant inhibition of DNA methyltransferase and BCL-2 protein function synergistically induce mitochondrial apoptosis in acute myelogenous leukemia cells. Ann Hematol. 91:1861–1870. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Garciaz S, Hospital MA, Alary AS, Saillard C, Hicheri Y, Mohty B, Rey J, D'Incan E, Charbonnier A, Villetard F, et al: Azacitidine plus venetoclax for the treatment of relapsed and newly diagnosed acute myeloid leukemia patients. Cancers. 14:20252022. View Article : Google Scholar : PubMed/NCBI | |
|
DiNardo CD, Pratz K, Pullarkat V, Jonas BA, Arellano M, Becker PS, Frankfurt O, Konopleva M, Wei AH, Kantarjian HM, et al: Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 133:7–17. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Jonas BA and Pollyea DA: How we use venetoclax with hypomethylating agents for the treatment of newly diagnosed patients with acute myeloid leukemia. Leukemia. 33:2795–2804. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Klepin HD: Definition of unfit for standard acute myeloid leukemia therapy. Curr Hematol Malig Rep. 11:537–544. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Senapati J, Kadia TM and Ravandi F: Maintenance therapy in acute myeloid leukemia: Advances and controversies. Haematologica. 108:2289–2304. 2023.PubMed/NCBI | |
|
Döhner H, Wei AH, Appelbaum FR, Craddock C, DiNardo CD, Dombret H, Ebert BL, Fenaux P, Godley LA, Hasserjian RP, et al: Diagnosis and management of AML in adults: 2022 recommendations from an international expert panel on behalf of the ELN. Blood. 140:1345–1377. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Schuurhuis GJ, Heuser M, Freeman S, Béné MC, Buccisano F, Cloos J, Grimwade D, Haferlach T, Hills RK, Hourigan CS, et al: Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood. 131:1275–1291. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Freites-Martinez A, Santana N, Arias-Santiago S and Viera A: Using the common terminology criteria for adverse events (CTCAE-Version 5.0) to evaluate the severity of adverse events of anticancer therapies. Actas Dermosifiliogr (Engl Ed). 112:90–92. 2021.(In English, Spanish). View Article : Google Scholar : PubMed/NCBI | |
|
DiNardo CD, Jonas BA, Pullarkat V, Thirman MJ, Garcia JS, Wei AH, Konopleva M, Döhner H, Letai A, Fenaux P, et al: Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 383:617–629. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Wei AH, Montesinos P, Ivanov V, DiNardo CD, Novak J, Laribi K, Kim I, Stevens DA, Fiedler W, Pagoni M, et al: Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: A phase 3 randomized placebo-controlled trial. Blood. 135:2137–2145. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pratz KW, Jonas BA, Pullarkat V, Recher C, Schuh AC, Thirman MJ, Garcia JS, DiNardo CD, Vorobyev V, Fracchiolla NS, et al: Measurable residual disease response and prognosis in Treatment-Naïve acute myeloid leukemia with venetoclax and azacitidine. J Clin Oncol. 40:855–865. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Chua CC, Hammond D, Kent A, Tiong IS, Konopleva MY, Pollyea DA, DiNardo CD and Wei AH: Treatment-free remission after ceasing venetoclax-based therapy in patients with acute myeloid leukemia. Blood Adv. 6:3879–3883. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Bernardi M, Ferrara F, Carrabba MG, Mastaglio S, Lorentino F, Vago L and Ciceri F: MRD in Venetoclax-based treatment for AML: Does it really matter? Front Oncol. 12:8908712022. View Article : Google Scholar : PubMed/NCBI | |
|
Alsouqi A, Geramita E and Im A: Treatment of acute myeloid leukemia in older adults. Cancers (Basel). 5:54092023. View Article : Google Scholar | |
|
Xin F, Yu YH, Shen XL and Zhang GX: The efficacy of the combination of venetoclax and hypomethylating agents versus HAG agents in patients with acute myeloid leukemia: A retrospective study. Hematology. 29:23503192024. View Article : Google Scholar : PubMed/NCBI | |
|
Rosko AE, Cordoba R, Abel G, Artz A, Loh KP and Klepin HD: Advances in management for older adults with hematologic malignancies. J Clin Oncol. 39:2102–2114. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Patel KK, Zeidan AM, Shallis RM, Prebet T, Podoltsev N and Huntington SF: Cost-effectiveness of azacitidine and venetoclax in unfit patients with previously untreated acute myeloid leukemia. Blood Adv. 5:994–1002. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Garciaz S, Dumas PY, Bertoli S, Sallman DA, Decroocq J, Belhabri A, Orvain C, Aspas Requena G, Simand C, Laribi K, et al: Outcomes of acute myeloid leukemia patients who responded to venetoclax and azacitidine and stopped treatment. Am J Hematol. 99:1870–1876. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Pollyea DA, DiNardo CD, Arellano ML, Pigneux A, Fiedler W, Konopleva M, Rizzieri DA, Smith BD, Shinagawa A, Lemoli RM, et al: Impact of venetoclax and azacitidine in Treatment-Naïve patients with acute myeloid leukemia and IDH1/2 mutations. Clin Cancer Res. 28:2753–2761. 2022. View Article : Google Scholar : PubMed/NCBI |