1,052 results on '"hypomethylating agents"'
Search Results
2. Oral decitabine/cedazuridine versus intravenous decitabine for acute myeloid leukaemia: A randomised, crossover, registration, pharmacokinetics study.
- Author
-
Geissler, Klaus, Koristek, Zdenek, del Castillo, Teresa Bernal, Novák, Jan, Rodríguez‐Macías, Gabriela, Metzelder, Stephan K., Illes, Arpad, Mayer, Jiří, Arnan, Montserrat, Keating, Mary‐Margaret, Krauter, Jürgen, Lunghi, Monia, Fracchiolla, Nicola Stefano, Platzbecker, Uwe, Santini, Valeria, Sano, Yuri, Oganesian, Aram, Keer, Harold, and Lübbert, Michael
- Abstract
Summary: This study compared decitabine exposure when administered IV (DEC‐IV) at a dose of 20 mg/m2 for 5‐days with orally administered decitabine with cedazuridine (DEC‐C), as well as the clinical efficacy and safety of DEC‐C in patients with acute myeloid leukaemia (AML) who were ineligible for intensive induction chemotherapy. In all, 89 patients were randomised 1:1 to DEC‐IV or oral DEC‐C (days 1–5 in a 28‐day treatment cycle), followed by 5 days of the other formulation in the next treatment cycle. All patients received oral DEC‐C for subsequent treatment cycles until treatment discontinuation. Equivalent systemic decitabine exposures were demonstrated (5‐day area under the curve ratio between the two decitabine formulations of 99.64 [90% confidence interval 91.23%, 108.80%]). Demethylation rates also were similar (≤1.1% difference). Median overall survival (OS), clinical response and safety profile with oral DEC‐C were consistent with those previously observed with DEC‐IV. Next‐generation sequencing was performed to identify molecular abnormalities that impact OS and TP53 mutations were associated with a poor outcome. These findings support the use of oral DEC‐C in patients with AML. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Soluble PD-L1 as a novel biomarker predicts poor outcomes and disease progression in de novo myelodysplastic syndromes.
- Author
-
Yang, Xingcheng, Jiang, Lijun, Zhang, Xiaoying, Peng, Juan, Qian, Hu, Huang, Lifang, He, Shaolong, Wang, Zhiqiong, Chen, Liting, Zhang, Yicheng, Ma, Ling, Chen, Yuan, and Wei, Jia
- Subjects
IMMUNE checkpoint proteins ,PROGRAMMED death-ligand 1 ,PROGRAMMED cell death 1 receptors ,OVERALL survival ,MYELODYSPLASTIC syndromes - Abstract
The role of the compromised immune microenvironment, including immune checkpoints, in myelodysplastic syndromes (MDS) has been identified as critical This study aimed to investigate the expression patterns of immune checkpoints, particularly soluble PD-1/PD-L1 (sPD-1/sPD-L1) as well as PD-1 on effector T cell subsets, and assess their prognostic value and potential regulatory roles in MDS. 161 MDS patients were enrolled, including 129 patients were primarily diagnosed with de novo MDS, together with 59 MDS patients who underwent hypomethylating agents (HMAs) therapy. Plasma sPD-L1 level was elevated in newly diagnosed MDS patients, which was also found to be associated with MDS disease progression that further increase in higher IPSS-R score group. Patients with increased sPD-L1 expression at diagnosis exhibited notably poorer overall survival, and multivariate Cox analysis indicated that elevated sPD-L1 was an independent risk factor. Furthermore, the levels of multiple cytokines and membrane-bound PD-1 on T cells were found to correlate with sPD-1/sPD-L1 levels in plasma. Importantly, we also found sPD-L1 levels significantly increased in MDS patients who showed progression of disease following HMAs therapy. In conclusion, we found elevated plasma sPD-L1 levels in MDS patients are associated with disease progression and poorer overall survival. This study showed that sPD-L1 is a potential biomarker for prognosis and a target for immunotherapy in MDS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Clinical efficacy and immune response of BCL-2 inhibitors combined with hypomethylating agents in the treatment of acute myeloid leukemia.
- Author
-
Peng, Xiaohuan, Yu, Jianing, Tang, Futian, Li, Yanhong, Bai, Jun, Li, Lijuan, and Zhang, Liansheng
- Subjects
LEUCOCYTES ,BONE marrow cells ,ERYTHROCYTES ,KILLER cells ,ACUTE myeloid leukemia ,CYTARABINE ,AZACITIDINE - Abstract
Objective: Acute myeloid leukemia (AML) is a malignant clonal proliferative disease with a high mortality rate. The combination therapy of BCL-2 inhibitor Venetoclax (VEN) and hypomethylating agents (HMAs) has significant anti-leukemia activity. Methods: We analyzed the efficacy, safety and immune response characteristics of AML patients who were unfit for high-dose chemotherapy and accepted the medication of VEN + HMAs. Results: After VEN + HMAs treatment, 31 newly diagnosed AML patients had the morphologic leukemia-free state rate (MLFS%) of 80.6% (25/31), complete response rate (CR%) of 54.8% (17/31), the minimal residual disease negative rate (MRD-%) of 51.6% (16/31), and the median progression-free survival (PFS) of 14 months. After treatment, the proportion of bone marrow primitive cells, the MRD level, white blood cell (WBC) count, fibrinogen (FIB) level and the proportion of B cells were significantly decreased. The red blood cell (RBC) count, hemoglobin (HGB) level, platelet count (PLT) count, activated partial thromboplastin time (APTT), the proportion of total T cells, CD8 + T cells and the IFN-γ level were significantly increased. After VEN + HMAs treatment, 12 relapsed AML patients had a MLFS% of 50% (6/12), CR% of 33.3% (4/12), MRD-% of 25% (3/12), and a median PFS of 7 months. After treatment, the proportion of bone marrow primitive cells and MRD level were slightly decreased, the proportions of CD8 + T cells and NK cells were significantly increased, the proportion of B cells and IL-10 level were significantly decreased. 12 AML patients who receive microtransplantation (MST) treatment using VEN + HMAs as a pretreatment regimen had a PFS of 20.5 months, which was much greater than VEN + HMAs group alone. Hematological recovery was better in the MST group with significantly increased RBC count, HGB level and PLT count. The most common adverse events were myelosuppression, agranulocytosis, infection and cardiovascular toxicity. No fatal adverse events were reported. Conclusion: The combination of BCL-2 inhibitors and HMAs had good efficacy and safety in AML patients who were unfit for high-dose chemotherapy, which may improve the immune microenvironment and enhance anti-leukemia immune response. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Decitabine-based treatment strategy improved the outcome of HSCT in JMML: a retrospective cohort study.
- Author
-
Zhiyong Peng, Jingyu Gao, Litao Huang, Yuelin He, Haoran Tang, Sa Zong, Yanru Pei, Fuyu Pei, Jing Ge, Xuan Liu, Li Yue, Jun Zhou, Xia Li, Dan Yue, Yun Chen, Chen Chen, Xuedong Wu, Xiaoqin Feng, and Chunfu Li
- Subjects
HEMATOPOIETIC stem cell transplantation ,CANCER chemotherapy ,OVERALL survival ,DISEASE remission ,PREVENTIVE medicine - Abstract
Introduction: Pre-HSCT disease control, suboptimal long-term prognosis, and a high recurrence incidence (RI) continue to pose significant challenges for hematopoietic stem cell transplantation (HSCT) in juvenilemyelomonocytic leukemia (JMML) patients. Methods: This retrospective cohort study assessed the effectiveness of a decitabine (DAC)-based protocol in JMML patients undergoing HSCT. The pre-HSCT treatment includes initial and bridging treatment. The efficacy of DACmonotherapy versus DAC combined with cytotoxic chemotherapy(C-DAC) as initial treatment was compared, followed by DAC plus FLAG (fludarabine, cytarabine, and GCSF) as bridging treatment. The HSCT regimens were based on DAC, fludarabine, and busulfan. Post-HSCT, low-dose DAC was used as maintenance therapy. The study endpoints focused on pretransplantation simplified clinical response and post-HSCT survival. Results: There were 109 patients, including 45 receiving DAC monotherapy and 64 undergoing C-DAC treatment. 106 patients completed bridging treatment. All patients were administered planned HSCT regimens and post-HSCT treatment. The initial treatment resulted in 88.1% of patients achieving clinical remission without a significant difference between the DAC and C-DAC groups (p=0.769). Clinical remission rates significantly improved following bridging treatment (p=0.019). The 5-year overall survival, leukemia-free survival, and RI were 92.2%, 88.4%, and 8.0%, respectively. A poor clinical response to pre-HSCT treatment emerged as a risk factor for OS (hazard ratio: 9.8, 95% CI: 2.3-41.1, p=0.002). Conclusion: Implementing a DAC-based administration strategy throughout the pre-HSCT period, during HSCT regimens, and in post-HSCT maintenance significantly reduced relapse and improved survival in JMML patients. Both DAC monotherapy and the DAC plus FLAG protocol proved effective as pre-HSCT treatments. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Clinical outcomes of hypomethylating agents and venetoclax in newly diagnosed unfit and relapsed/refractory paediatric, adolescent and young adult acute myeloid leukaemia patients.
- Author
-
LeBlanc, Francis R., Breese, Erin H., Burns, Karen C., Chang, Ellen K., Jones, LaQuita M., Lee, Lynn, Mizukawa, Benjamin, Norris, Robin E., O'Brien, Maureen M., Phillips, Christine L., Perentesis, John P., Rubinstein, Jeremy, and Pommert, Lauren
- Subjects
- *
HEMATOPOIETIC stem cell transplantation , *ACUTE myeloid leukemia , *YOUNG adults , *DISEASE relapse , *CHILDREN'S hospitals - Abstract
Summary: Venetoclax (VEN) combined with hypomethylating agents (HMA) decitabine or azacitidine is used for adult acute myeloid leukaemia (AML), but its application in paediatric, adolescent and young adult (AYA) AML lacks prospective studies. We performed a retrospective chart review of paediatric and AYA AML patients treated with HMA + VEN at Cincinnati Children's Hospital Medical Centre. Twenty‐seven patients received 30 HMA + VEN treatment courses for relapsed/refractory (R/R, n = 21) or newly diagnosed (n = 9) AML due to ineligibility for intensive chemotherapy. The R/R cohort had high‐risk cytomolecular genetic alterations and prior extensive treatments, with 50% (n = 9) of relapse patients (n = 18) having undergone haematopoietic stem cell transplantation (HSCT). Venetoclax treatment using the 400 mg adult exposure‐equivelant dosing (AED) had a median duration of 21 days (range 7–30 days). Grade 3–4 toxicities included neutropenia (90%), anaemia (64%), thrombocytopenia (64%) and febrile neutropenia (44%). The overall complete remission (CR)/CR with incomplete blood count recovery (CRi) rate was 73% (77% minimal residual disease [MRD] negativity <0.1%), with 60% undergoing HSCT. Among newly diagnosed patients (n = 9), 89% achieved CR/CRi (78% MRD negativity) and 78% proceeded to HSCT. The R/R cohort (n = 21) showed a 67% CR/CRi rate (71% MRD negativity), with 52% undergoing HSCT. These findings support the safety and efficacy of HMA + VEN in paediatric/AYA AML, indicating it as a viable option for patients unfit for intensive chemotherapy. Further studies are necessary to determine optimal venetoclax dosing, chemotherapy combinations and pharmacokinetics in this population. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Advances in the management of higher-risk myelodysplastic syndromes: future prospects.
- Author
-
Gener-Ricos, Georgina, Rodriguez-Sevilla, Juan Jose, Urrutia, Samuel, Bataller, Alex, Bazinet, Alexandre, and Garcia-Manero, Guillermo
- Subjects
- *
HEMATOPOIETIC stem cell transplantation , *MYELODYSPLASTIC syndromes , *STEM cell transplantation , *BONE marrow , *AZACITIDINE - Abstract
Higher-risk myelodysplastic syndromes (HR-MDS) are defined using a number of prognostic scoring systems that include the degree of cytopenias, percentage of blasts, cytogenetic alterations, and more recently genomic data. HR-MDS encompasses characteristics such as progressive cytopenias, increased bone marrow blasts, unfavorable cytogenetics, and an adverse mutational profile. Survival is generally poor, and patients require therapy to improve outcomes. Hypomethylating agents (HMAs), such as azacitidine, decitabine, and more recently, oral decitabine/cedazuridine, are the only approved therapies for HR-MDS. These are often continued until loss of response, progression, or unacceptable toxicity. Combinations including an HMA plus other drugs have been investigated but have not demonstrated better outcomes compared to single-agent HMA. Moreover, in a disease of high genomic complexity such as HR-MDS, therapy targeting specific genomic abnormalities is of interest. This review will examine the biological underpinnings of HR-MDS, its therapeutic landscape in the frontline and relapsed settings, as well as the impact of hematopoietic stem cell transplantation, the only known curative intervention for this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Soluble PD-L1 as a novel biomarker predicts poor outcomes and disease progression in de novo myelodysplastic syndromes
- Author
-
Xingcheng Yang, Lijun Jiang, Xiaoying Zhang, Juan Peng, Hu Qian, Lifang Huang, Shaolong He, Zhiqiong Wang, Liting Chen, Yicheng Zhang, Ling Ma, Yuan Chen, and Jia Wei
- Subjects
Myelodysplastic syndromes ,PD-1/PD-L1 signaling pathway ,Soluble immune checkpoints ,Cytokines ,Hypomethylating agents ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract The role of the compromised immune microenvironment, including immune checkpoints, in myelodysplastic syndromes (MDS) has been identified as critical This study aimed to investigate the expression patterns of immune checkpoints, particularly soluble PD-1/PD-L1 (sPD-1/sPD-L1) as well as PD-1 on effector T cell subsets, and assess their prognostic value and potential regulatory roles in MDS. 161 MDS patients were enrolled, including 129 patients were primarily diagnosed with de novo MDS, together with 59 MDS patients who underwent hypomethylating agents (HMAs) therapy. Plasma sPD-L1 level was elevated in newly diagnosed MDS patients, which was also found to be associated with MDS disease progression that further increase in higher IPSS-R score group. Patients with increased sPD-L1 expression at diagnosis exhibited notably poorer overall survival, and multivariate Cox analysis indicated that elevated sPD-L1 was an independent risk factor. Furthermore, the levels of multiple cytokines and membrane-bound PD-1 on T cells were found to correlate with sPD-1/sPD-L1 levels in plasma. Importantly, we also found sPD-L1 levels significantly increased in MDS patients who showed progression of disease following HMAs therapy. In conclusion, we found elevated plasma sPD-L1 levels in MDS patients are associated with disease progression and poorer overall survival. This study showed that sPD-L1 is a potential biomarker for prognosis and a target for immunotherapy in MDS.
- Published
- 2024
- Full Text
- View/download PDF
9. Clinical efficacy and immune response of BCL-2 inhibitors combined with hypomethylating agents in the treatment of acute myeloid leukemia
- Author
-
Xiaohuan Peng, Jianing Yu, Futian Tang, Yanhong Li, Jun Bai, Lijuan Li, and Liansheng Zhang
- Subjects
Acute myeloid leukemia ,Clinical efficacy ,Immune response ,BCL-2 inhibitors ,Hypomethylating agents ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Objective Acute myeloid leukemia (AML) is a malignant clonal proliferative disease with a high mortality rate. The combination therapy of BCL-2 inhibitor Venetoclax (VEN) and hypomethylating agents (HMAs) has significant anti-leukemia activity. Methods We analyzed the efficacy, safety and immune response characteristics of AML patients who were unfit for high-dose chemotherapy and accepted the medication of VEN + HMAs. Results After VEN + HMAs treatment, 31 newly diagnosed AML patients had the morphologic leukemia-free state rate (MLFS%) of 80.6% (25/31), complete response rate (CR%) of 54.8% (17/31), the minimal residual disease negative rate (MRD-%) of 51.6% (16/31), and the median progression-free survival (PFS) of 14 months. After treatment, the proportion of bone marrow primitive cells, the MRD level, white blood cell (WBC) count, fibrinogen (FIB) level and the proportion of B cells were significantly decreased. The red blood cell (RBC) count, hemoglobin (HGB) level, platelet count (PLT) count, activated partial thromboplastin time (APTT), the proportion of total T cells, CD8 + T cells and the IFN-γ level were significantly increased. After VEN + HMAs treatment, 12 relapsed AML patients had a MLFS% of 50% (6/12), CR% of 33.3% (4/12), MRD-% of 25% (3/12), and a median PFS of 7 months. After treatment, the proportion of bone marrow primitive cells and MRD level were slightly decreased, the proportions of CD8 + T cells and NK cells were significantly increased, the proportion of B cells and IL-10 level were significantly decreased. 12 AML patients who receive microtransplantation (MST) treatment using VEN + HMAs as a pretreatment regimen had a PFS of 20.5 months, which was much greater than VEN + HMAs group alone. Hematological recovery was better in the MST group with significantly increased RBC count, HGB level and PLT count. The most common adverse events were myelosuppression, agranulocytosis, infection and cardiovascular toxicity. No fatal adverse events were reported. Conclusion The combination of BCL-2 inhibitors and HMAs had good efficacy and safety in AML patients who were unfit for high-dose chemotherapy, which may improve the immune microenvironment and enhance anti-leukemia immune response.
- Published
- 2024
- Full Text
- View/download PDF
10. Real‐world study of the use of azacitidine in myelodysplasia in Australia
- Author
-
Anoop Enjeti, Asma Ashraf, Vincent Caillet, Arif Alam, Jonathan Silar, Harold Keer, Francesco Castaldi, and Taleisha Paine
- Subjects
azacitidine ,hypomethylating agents ,myelodysplastic syndromes ,real‐world ,survival ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract Hypomethylating agents are the most widely used upfront therapy for patients with myelodysplastic syndrome (MDS) who are not suitable for hematopoietic stem cell transplantation. In Australia, azacitidine was, until recently, the only approved and subsidized treatment for patients with intermediate‐2 and high‐risk MDS, chronic myelomonocytic leukemia, and low blast acute myeloid leukemia. We analyzed prescription data to evaluate the real‐world persistence and overall survival (OS) of patients prescribed azacitidine for the first time in Australia. A retrospective cohort analysis of patients who had been prescribed Pharmaceutical Benefits Scheme (PBS)‐listed azacitidine for the first time, between January 2016 and April 2021, was conducted using the PBS 10% dataset. Treatment persistence and OS were estimated using Kaplan–Meier methods. The impact of the number of treatment cycles and treatment adherence on OS was also estimated. There were 351 patients in the PBS 10% dataset who initiated treatment with azacitidine. The average age (standard deviation [SD]) at azacitidine initiation was 71.9 (11.1) years and the average number (SD) of azacitidine prescriptions was 5.6 (0.2). The median persistence on azacitidine was 15.6 months, and the OS was 13.4 months. The median OS for patients who had six or more cycles of azacitidine treatment was greater compared to patients who had five or less cycles of treatment. The data from this real‐world study illustrate the unmet medical needs of patients with MDS treated with azacitidine in Australia. The majority of patients are not treated with the optimal number of cycles of azacitidine, which is negatively correlated with patient outcomes.
- Published
- 2024
- Full Text
- View/download PDF
11. Development of the treatment preference in myelodysplasia questionnaire for clinicians, carers, and patients
- Author
-
Robert Morlock, Chun Fong, Francesco Castaldi, Taliesha Paine, Donna Collett, and Anoop Enjeti
- Subjects
hypomethylating agents ,myelodysplastic syndromes ,questionnaires ,treatment preference ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Abstract This study reports the development activities for the Treatment Preference Myelodysplasia Questionnaires (TPMQ) for clinicians (mTPMQ), carers (cTPMQ), and patients (pTPMQ). These tools are intended to evaluate treatment preferences for patients with myelodysplastic syndromes (MDS). This was a non‐interventional, cross‐sectional qualitative interview study consisting of interviews with clinicians, patients, and those caring for patients with MDS. All participants were located in Australia and data were collected from qualitative mixed‐method interviews composed of concept elicitation and cognitive debriefing related to initial drafts of the questionnaires. Fifteen individuals participated in interviews (five from each group). Based on the concept elicitation portion of interviews, concepts of importance were classified and reasons for treatment preference were documented. From cognitive debriefing, the questionnaires were generally deemed to be clear and easy to understand. Participant input from both concept elicitation and cognitive debriefing portions was used to revise initial drafts of the questionnaires. The mTPMQ, cTPMQ, and pTPMQ were developed with direct input from clinicians, patients, and caregivers to assess the key concepts of interest related to the preference for the treatment of MDS and are ready to be used and evaluated further in clinical trials.
- Published
- 2024
- Full Text
- View/download PDF
12. Decitabine cytotoxicity is promoted by dCMP deaminase DCTD and mitigated by SUMO-dependent E3 ligase TOPORS.
- Author
-
Carnie, Christopher J, Götz, Maximilian J, Palma-Chaundler, Chloe S, Weickert, Pedro, Wanders, Amy, Serrano-Benitez, Almudena, Li, Hao-Yi, Gupta, Vipul, Awwad, Samah W, Blum, Christian J, Sczaniecka-Clift, Matylda, Cordes, Jacqueline, Zagnoli-Vieira, Guido, D'Alessandro, Giuseppina, Richards, Sean L, Gueorguieva, Nadia, Lam, Simon, Beli, Petra, Stingele, Julian, and Jackson, Stephen P
- Subjects
- *
CYTOTOXINS , *UBIQUITIN ligases , *GENETIC testing , *DECITABINE , *DNA methyltransferases , *UBIQUITINATION - Abstract
The nucleoside analogue decitabine (or 5-aza-dC) is used to treat several haematological cancers. Upon its triphosphorylation and incorporation into DNA, 5-aza-dC induces covalent DNA methyltransferase 1 DNA–protein crosslinks (DNMT1-DPCs), leading to DNA hypomethylation. However, 5-aza-dC's clinical outcomes vary, and relapse is common. Using genome-scale CRISPR/Cas9 screens, we map factors determining 5-aza-dC sensitivity. Unexpectedly, we find that loss of the dCMP deaminase DCTD causes 5-aza-dC resistance, suggesting that 5-aza-dUMP generation is cytotoxic. Combining results from a subsequent genetic screen in DCTD-deficient cells with the identification of the DNMT1-DPC-proximal proteome, we uncover the ubiquitin and SUMO1 E3 ligase, TOPORS, as a new DPC repair factor. TOPORS is recruited to SUMOylated DNMT1-DPCs and promotes their degradation. Our study suggests that 5-aza-dC-induced DPCs cause cytotoxicity when DPC repair is compromised, while cytotoxicity in wild-type cells arises from perturbed nucleotide metabolism, potentially laying the foundations for future identification of predictive biomarkers for decitabine treatment. Synopsis: The chemotherapeutic decitabine/5-aza-dC induces covalent DNMT1 DNA-protein crosslinks (DPCs), leading to DNMT1 degradation and DNA hypomethylation. This study finds that E3 ligase TOPORS promotes DNMT1-DPC degradation, while deaminase DCTD mediates a DNMT1-independent mode of 5-aza-dC cytotoxicity. The dCMP deaminase DCTD mediates 5-aza-dC cytotoxicity independently of DNA-protein crosslink formation, through the generation of 5-aza-dUMP. The ubiquitin and SUMO1 E3 ligase TOPORS is a novel player in global-genome DPC repair. TOPORS is recruited to SUMOylated DNMT1-DPCs through its SUMO-interaction motifs, whereupon it promotes DNMT1-DPC ubiquitylation and degradation. TOPORS acts in parallel to the E3 ubiquitin ligase RNF4 in mediating DNMT1-DPC tolerance. TOPORS acts in parallel to the ubiquitin ligase RNF4 in degradation of DNMT1-DNA crosslinks, thereby mediating cellular tolerance to these DPCs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Clinical activity, pharmacokinetics, and pharmacodynamics of oral hypomethylating agents for myelodysplastic syndromes/neoplasms and acute myeloid leukemia: A multidisciplinary review.
- Author
-
Haumschild, Ryan, Kennerly-Shah, Julie, Barbarotta, Lisa, and Zeidan, Amer M.
- Subjects
- *
MYELODYSPLASTIC syndromes , *PATIENT selection , *DRUG toxicity , *PATIENT compliance , *HEMATOLOGIC malignancies , *PATIENT safety , *ANTINEOPLASTIC agents , *AZACITIDINE , *ORAL drug administration , *SYMPTOMS , *CANCER patients , *TREATMENT effectiveness , *PARENTERAL infusions , *INFORMATION needs , *ATTITUDES of medical personnel , *DECITABINE , *QUALITY of life , *DRUGS , *QUALITY assurance , *HEALTH care teams , *PHARMACODYNAMICS - Abstract
Objective: To review the pharmacokinetic (PK)–pharmacodynamic (PD) profiles, disease setting, dosing, and safety of oral and parenteral hypomethylating agents (HMAs) for the treatment of myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukemia (AML), and to provide a multidisciplinary perspective on treatment selection and educational needs relating to HMA use. Data Sources: Clinical and real-world data for parenteral decitabine and azacitidine and two oral HMAs: decitabine–cedazuridine (DEC-C) for MDS and azacitidine (CC-486) for AML maintenance therapy. Data Summary: Differences in the PK–PD profiles of oral and parenteral HMA formulations have implications for their potential toxicities and planned use. Oral DEC-C (decitabine 35 mg and cedazuridine 100 mg) has demonstrated equivalent systemic area under the concentration-time curve (AUC) exposure to a 5-day regimen of intravenous (IV) decitabine 20 mg/m2 and showed no significant difference in PD. The AUC equivalence of oral DEC-C and IV decitabine means that these regimens can be treated interchangeably (but must not be substituted within a cycle). Oral azacitidine has a distinct PK–PD profile versus IV or subcutaneous azacitidine, and the formulations are not bioequivalent or interchangeable owing to differences in plasma time-course kinetics and exposures. Clinical trials are ongoing to evaluate oral HMA combinations and novel oral HMAs, such as NTX-301 and ASTX030. Conclusions: Treatment with oral HMAs has the potential to improve quality of life, treatment adherence, and disease outcomes versus parenteral HMAs. Better education of multidisciplinary teams on the factors affecting HMA treatment selection may help to improve treatment outcomes in patients with MDS or AML. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Entospletinib with decitabine in acute myeloid leukemia with mutant TP53 or complex karyotype: A phase 2 substudy of the Beat AML Master Trial
- Author
-
Duong, Vu H, Ruppert, Amy S, Mims, Alice S, Borate, Uma, Stein, Eytan M, Baer, Maria R, Stock, Wendy, Kovacsovics, Tibor, Blum, William, Arellano, Martha L, Schiller, Gary J, Olin, Rebecca L, Foran, James M, Litzow, Mark R, Lin, Tara L, Patel, Prapti A, Foster, Matthew C, Redner, Robert L, Al‐Mansour, Zeina, Cogle, Christopher R, Swords, Ronan T, Collins, Robert H, Vergilio, Jo‐Anne, Heerema, Nyla A, Rosenberg, Leonard, Yocum, Ashley O, Marcus, Sonja, Chen, Timothy, Druggan, Franchesca, Stefanos, Mona, Gana, Theophilus J, Shoben, Abigail B, Druker, Brian J, Burd, Amy, Byrd, John C, Levine, Ross L, and Boyiadzis, Michael M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Clinical Trials and Supportive Activities ,Hematology ,Pediatric ,Rare Diseases ,Childhood Leukemia ,Clinical Research ,Cancer ,Orphan Drug ,Aging ,Pediatric Cancer ,6.1 Pharmaceuticals ,Humans ,Decitabine ,Tumor Suppressor Protein p53 ,Leukemia ,Myeloid ,Acute ,Karyotype ,Treatment Outcome ,Antineoplastic Combined Chemotherapy Protocols ,acute myeloid leukemia ,decitabine ,entospletinib ,hypomethylating agents ,tumor protein p53 ,Public Health and Health Services ,Oncology & Carcinogenesis ,Oncology and carcinogenesis ,Public health - Abstract
BackgroundPatients with acute myeloid leukemia (AML) who have tumor protein p53 (TP53) mutations or a complex karyotype have a poor prognosis, and hypomethylating agents are often used. The authors evaluated the efficacy of entospletinib, an oral inhibitor of spleen tyrosine kinase, combined with decitabine in this patient population.MethodsThis was a multicenter, open-label, phase 2 substudy of the Beat AML Master Trial (ClinicalTrials.gov identifier NCT03013998) using a Simon two-stage design. Eligible patients aged 60 years or older who had newly diagnosed AML with mutations in TP53 with or without a complex karyotype (cohort A; n = 45) or had a complex karyotype without TP53 mutation (cohort B; n = 13) received entospletinib 400 mg twice daily with decitabine 20 mg/m2 on days 1-10 every 28 days for up to three induction cycles, followed by up to 11 consolidation cycles, in which decitabine was reduced to days 1-5. Entospletinib maintenance was given for up to 2 years. The primary end point was complete remission (CR) and CR with hematologic improvement by up to six cycles of therapy.ResultsThe composite CR rates for cohorts A and B were 13.3% (95% confidence interval, 5.1%-26.8%) and 30.8% (95% confidence interval, 9.1%-61.4%), respectively. The median duration of response was 7.6 and 8.2 months, respectively, and the median overall survival was 6.5 and 11.5 months, respectively. The study was stopped because the futility boundary was crossed in both cohorts.ConclusionsThe combination of entospletinib and decitabine demonstrated activity and was acceptably tolerated in this patient population; however, the CR rates were low, and overall survival was short. Novel treatment strategies for older patients with TP53 mutations and complex karyotype remain an urgent need.
- Published
- 2023
15. Hypomethylating agent-based therapies in older adults with acute myeloid leukemia - A joint review by the Young International Society of Geriatric Oncology and European Society for Blood and Marrow Transplantation Trainee Committee.
- Author
-
Neuendorff, Nina, Gagelmann, Nico, Meckstroth, Shelby, Thibaud, Vincent, Zhao, Yue, Mir, Nabiel, Shih, Yung-Yu, Amaro, Danielle, Roy, Mukul, Lombardo, Joseph, Gjærde, Lars, Loh, Kah, and Singhal, Surbhi
- Subjects
Acute myeloid leukemia ,Azacitidine ,Decitabine ,Guadecitabine ,Hypomethylating agents ,Older adults ,Humans ,Aged ,Decitabine ,Bone Marrow ,Quality of Life ,Antineoplastic Agents ,Azacitidine ,Leukemia ,Myeloid ,Acute ,Antineoplastic Combined Chemotherapy Protocols - Abstract
Acute myeloid leukemia (AML) is associated with poor outcomes in older adults. A major goal of treatment is to balance quality of life and functional independence with disease control. With the approval of new, more tolerable regimens, more older adults are able to receive AML-directed therapy. Among these options are hypomethylating agents (HMAs), specifically azacitidine and decitabine. HMAs have become an integral part of AML therapy over the last two decades. These agents are used either as monotherapy or nowadays more commonly in combination with other agents such as the Bcl-2 inhibitor venetoclax. Biological AML characteristics, such as molecular and cytogenetic risk factors, play crucial roles in guiding treatment decisions. In patients with high-risk AML, HMAs are increasingly used rather than intensive chemotherapy, although further trials based on a risk-adapted approach using patient- and disease-related factors are needed. Here, we review trials and evidence for the use of HMA monotherapy and combination therapy in the management of older adults with AML. Furthermore, we discuss the use of HMAs and HMA combination therapies in AML, mechanisms of action, their incorporation into hematopoietic stem cell transplantation strategies, and their use in patients with comorbidities and reduced organ function.
- Published
- 2023
16. Evaluation of infectious complications in patients with myelodysplastic syndromes: a prospective cohort study from the Canadian MDS registry
- Author
-
Mathur, S., Christou, G., Delage, R., Elemary, M., Finn, N., Geddes, M., Houston, DS., Keating, MM., Khalaf, D., Leber, B., Leitch, H., Lother, SA., Mozessohn, L., Nevill, T., Parmentier, A., Paulson, K., Rimmer, E., Sabloff, M., Shamy, A., St-Hilaire, E., Storring, J., Yee, K., Zhang, L., Zhu, N., Hay, AE., Zarychanski, R., Buckstein, R., and Houston, Brett L.
- Published
- 2024
- Full Text
- View/download PDF
17. AML in the Elderly – When less may be more
- Author
-
Winer, Eric S. and Stone, Richard M.
- Published
- 2024
- Full Text
- View/download PDF
18. Successfully treatment of PLZF-RARα positive acute promyelocytic leukemia by Venetoclax combining with decitabine: a case report and literature review
- Author
-
Ziwei Zhou, Erling Chen, Jiqian Jiang, Lei Xue, Lijun Zhu, Xing Hu, Yingqiao Zhu, Changcheng Zheng, and Juan Tong
- Subjects
Acute promyelocytic leukemia (APL) ,case report ,PLZF-RARα ,Venetoclax ,t(11, 17) (q23 ,q21) ,hypomethylating agents ,Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Introduction: Acute promyelocytic leukemia (APL) is mainly due to the chromosome translocation t(15; 17) (q22; q12), leading to the formation of PML-RARα fusion protein. However, some patients carried rare translocation involving RARα gene, and they were referred to as variant APL caused by the RAR family (RARα, RARB, and RARG) and partner genes. PLZF-RARα was a rare type of molecular genetic abnormality with unfavorable prognosis that has been reported in few cases in variant APL. Knowledge of PLZF-RARα (+) APL treatment remains limited understood.Case report: We presented a case of variant APL in a 47-year-old female, who was PLZF-RARα positive detected by reverse transcription polymerase chain reaction (RT–PCR). The patient did not respond to all-trans retinoic acid (ATRA), idarubicin, and arsenic trioxide (As2O3) combined induction chemotherapy. Then, the patient was treated with Venetoclax combining with decitabine as the salvage therapy and achieved morphological remission and PLZF/RARα gene negative.Conclusion: Venetoclax combining with decitabine can be used as an effective therapy in the PLZF-RARα positive APL.
- Published
- 2024
- Full Text
- View/download PDF
19. Ex vivo characterization of acute myeloid leukemia patients undergoing hypomethylating agents and venetoclax regimen reveals a venetoclax-specific effect on non-suppressive regulatory T cells and bona fide PD-1+TIM3+ exhausted CD8+ T cells.
- Author
-
Corradi, Giulia, Forte, Dorian, Cristiano, Gianluca, Polimeno, Andrea, Ciciarello, Marilena, Salvestrini, Valentina, Bandini, Lorenza, Robustelli, Valentina, Ottaviani, Emanuela, Cavo, Michele, Ocadlikova, Darina, and Curti, Antonio
- Subjects
REGULATORY T cells ,CYTARABINE ,ACUTE myeloid leukemia ,VENETOCLAX ,IMMUNOTHERAPY ,T cells ,IMMUNE checkpoint proteins - Abstract
Acute myeloid leukemia (AML) is an aggressive heterogeneous disease characterized by several alterations of the immune system prompting disease progression and treatment response. The therapies available for AML can affect lymphocyte function, limiting the efficacy of immunotherapy while hindering leukemia-specific immune reactions. Recently, the treatment based on Venetoclax (VEN), a specific B-cell lymphoma 2 (BCL-2) inhibitor, in combination with hypomethylating agents (HMAs) or low-dose cytarabine, has emerged as a promising clinical strategy in AML. To better understand the immunological effect of VEN treatment, we characterized the phenotype and immune checkpoint (IC) receptors' expression on CD4
+ and CD8+ T cells from AML patients after the first and second cycle of HMA in combination with VEN. HMA and VEN treatment significantly increased the percentage of naive CD8+ T cells and TIM-3+ CD4+ and CD8+ T cells and reduced cytokine-secreting non-suppressive T regulatory cells (Tregs). Of note, a comparison between AML patients treated with HMA only and HMA in combination with VEN revealed the specific contribution of VEN in modulating the immune cell repertoire. Indeed, the reduction of cytokine-secreting non-suppressive Tregs, the increased TIM-3 expression on CD8+ T cells, and the reduced co-expression of PD-1 and TIM-3 on both CD4+ and CD8+ T cells are all VEN-specific. Collectively, our study shed light on immune modulation induced by VEN treatment, providing the rationale for a novel therapeutic combination of VEN and IC inhibitors in AML patients. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
20. A real-world experience of venetoclax combined with hypomethylating agents vs. monotherapy hypomethylating agents in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia patients.
- Author
-
Ludan Zhang, Rui Ge, Deng Pan, Pengjie Yue, Jingwen Zhang, Renjie Bian, and Xiaojing Yan
- Subjects
CHRONIC leukemia ,MYELODYSPLASTIC syndromes ,VENETOCLAX ,OVERALL survival ,PROGRESSION-free survival - Abstract
Introduction: Current clinical research has reported the effectiveness and safety of venetoclax in combination with hypomethylating agents (VEN-HMA) in patients with myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). Thus, this study aimed to examine the effectiveness and safety of VEN-HMA therapy in patients with MDS and CMML and compared its shortterm and long-term therapeutic effects with HMA monotherapy. Method: We analyzed data from our center, comprising 19 patients with MDS and CMML who received VEN-HMA therapy, compared to 32 patients treated with HMA monotherapy. Results: The overall response rate (ORR) in the VEN-HMA group was 73.7%, compared to 59.4% in the HMA group. The survival analysis revealed that the median overall survival (mOS) time in the VEN-HMA group was 16 months, with a median progression-free survival (mPFS) time of 9 months, both of which were longer than those observed in the HMA group (p < 0.05). Key adverse events (AEs) included grade 3-4 neutropenia (89.5% in VEN-HMA group vs. 87.5% in HMA group), grade 3-4 thrombocytopenia (73.7% vs. 71.9%), and anemia (73.7% vs. 90.6%). Infection of grade 3 or higher occurred in 63.2% of patients in the VENHMA group and 65.6% of patients in the HMA group. Discussion: Our study has confirmed the effectiveness and safety of the combined treatment of HMAs and venetoclax, which offers significant advantages to patients due to the relatively high and rapid response rates. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
21. DNA methylation profiling of myelodysplastic syndromes and clinical response to azacitidine: A multicentre retrospective study.
- Author
-
Noguera‐Castells, Aleix, Campillo‐Marcos, Ignacio, Davalos, Veronica, García‐Prieto, Carlos A., Valcárcel, David, Molero, Antonieta, Palomo, Laura, Gattermann, Norbert, Wulfert, Michael, Chaparro‐González, Lorea, Solé, Francesc, Cabezón, Marta, Jiménez‐Lorenzo, María J., Xicoy, Blanca, Zamora, Lurdes, De Stefano, Alessia, Casalin, Irene, Finelli, Carlo, Follo, Matilde Y., and Esteller, Manel
- Subjects
- *
MYELODYSPLASTIC syndromes , *DNA methylation , *AZACITIDINE , *RETROSPECTIVE studies , *EPIGENOMICS , *BIOMARKERS - Abstract
Summary: Real‐world data have revealed that a substantial portion of patients with myelodysplastic syndromes (MDS) does not respond to epigenetic therapy with hypomethylating agents (HMAs). The cellular and molecular reasons for this resistance to the demethylating agent and biomarkers that would be able to predict the treatment refractoriness are largely unknown. In this study, we shed light on this enigma by characterizing the epigenomic profiles of patients with MDS treated with azacitidine. Our approach provides a comprehensive view of the evolving DNA methylation architecture of the disease and holds great potential for advancing our understanding of MDS treatment responses to HMAs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Donor Lymphocyte Infusion Is a Feasible Way to Improve Survival in Patients with Acute Myeloid Leukemia and Myelodysplastic Syndromes Who Relapse after Allogeneic Stem Cell Transplantation.
- Author
-
Minculescu, Lia, Reekie, Joanne, Petersen, Soeren Lykke, Kornblit, Brian Thomas, Schjoedt, Ida, Andersen, Niels Smedegaard, Andersen, Lisbeth Pernille, Fischer-Nielsen, Anne, Haastrup, Eva Kannik, Friis, Lone Smidstrup, and Sengelov, Henrik
- Subjects
- *
STEM cell transplantation , *ACUTE myeloid leukemia , *MYELODYSPLASTIC syndromes , *OVERALL survival , *LYMPHOCYTES - Abstract
Introduction: Donor lymphocyte infusion (DLI) is used to induce remission in patients who relapse after allogeneic stem cell transplantation (allo-HSCT). During the last decade, the hypomethylating agent Azacitidine has been used together with DLI for a synergistic graft-versus-leukemia (GVL) effect. Here, we report results of DLI/Azacitidine treatment from a retrospective single-center study. Methods: Fifty AML/MDS patients treated for relapse after allo-HSCT between 2001 and 2020 with DLI at the Department of Hematology, at Rigshospitalet, Copenhagen University Hospital were included for analyses. A subgroup of patients who obtained complete remission (CR) after reinduction chemotherapy, received DLI in combination with low-dose (32 mg/m2) Azacitidine. Results: Overall survival in all patients after DLI treatment was 59% at 2 years and 20% at 5 years. Relapse-free survival in patients in CR prior to DLI was 32% after 2 years and 7% after 5 years. In the DLI + low-dose-Azacitidine group, 5-year relapse-free survival was 40%. Conclusion: DLI remains an effective treatment in post-transplant relapse leaving one-fifth of patients' long-term survivors. Our results support the concomitant use of low-dose Azacitidine in the future use of DLI in order to enhance the GVL effect of donor lymphocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
23. Efficacy and safety of venetoclax plus hypomethylating agents in relapsed/refractory acute myeloid leukemia: a multicenter real-life experience.
- Author
-
Angotzi, Francesco, Lessi, Federica, Leoncin, Matteo, Filì, Carla, Endri, Mauro, Lico, Albana, Visentin, Andrea, Pravato, Stefano, Candoni, Anna, Trentin, Livio, and Gurrieri, Carmela
- Subjects
ACUTE myeloid leukemia ,VENETOCLAX ,STEM cell transplantation ,PROGNOSIS ,SURVIVAL rate - Abstract
Venetoclax (VEN) has been shown to play a synergistic effect in combination with hypomethylating agents (HMAs) in the frontline treatment of acute myeloid leukemia (AML). However, the potential role of this therapy in the relapsed/refractory (R/R) AML setting, still needs to be further unveiled. The aim of the current study was to retrospectively outline the safety profile, response and survival outcomes of R/R AML patients treated with VEN in association with HMAs. Clinical, biological, and molecular data were collected from 57 patients with R/R AML treated with VEN combined with azacitidine or decitabine between 2018 and 2023. The median age of patients was 63 years, 38 (66.7%) received treatment for relapsed disease while 19 (33.3%) for refractory disease, 5 (8.7%) were treated for molecular relapse. A consistent proportion of the cohort was represented by patients with unfavorable prognostic factors such as complex karyotype (36.8%), secondary AML (29.8%), previous exposure to HMAs (38.6%), and relapse after allogeneic stem cell transplant (22.8%). A total of 14 patients achieved CR (24.6%), 3 (5.3%) CRi, 3 (5.3%) MLFS, and 3 (5.3%) PR, accounting for an ORR of 40.4%. The CR/CRi rate was higher in the group treated with azacitidine than in the group treated with decitabine (37.8% vs. 15%). The median OS was 8.2 months, reaching 20.1 months among responding patients. VEN-HMAs treatment allowed to bridge to allogeneic stem cell transplantation 11 (23.9%) of eligible patients, for which a median OS of 19.8 months was shown. On multivariate analysis, ECOG performance status ≥2, complex karyotype and not proceeding to allogeneic stem cell transplantation after therapy with VEN-HMAs were the factors independently associated with shorter OS. Patients treated with the azacitidine rather than the decitabine containing regimen generally displayed a trend toward superior outcomes. The major toxicities were prolonged neutropenia and infections. In conclusion, this study showed how VEN-HMAs could represent an effective salvage therapy in patients with R/R AML, even among some of those patients harboring dismal prognostic features, with a good toxicity profile. Further prospective studies are thus warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
24. Latest Insights and Therapeutic Advances in Myelodysplastic Neoplasms.
- Author
-
Niscola, Pasquale, Gianfelici, Valentina, Giovannini, Marco, Piccioni, Daniela, Mazzone, Carla, and de Fabritiis, Paolo
- Subjects
- *
MYELODYSPLASTIC syndromes treatment , *MYELODYSPLASTIC syndromes , *HEMATOLOGIC malignancies , *DISEASE risk factors - Abstract
Simple Summary: Several recent studies have demonstrated encouraging results in treating patients with myelodysplastic syndromes/neoplasms (MDSs). This review focuses on the latest conceptual and therapeutic advances in the management of adults with MDS, addressing diagnostic principles, classification updates, and prognostic stratification systems, as well as improvements in clinical approaches that provide significant benefits through novel treatments, which may, in turn, represent a valuable basis for developing future clinical trials. Myelodysplastic syndromes/neoplasms (MDSs) encompass a range of hematopoietic malignancies, commonly affecting elderly individuals. Molecular alterations in the hematopoietic stem cell compartment drive disease pathogenesis. Recent advancements in genomic profiling have provided valuable insights into the biological underpinnings of MDSs and have expanded therapeutic options, particularly for specific molecularly defined subgroups. This review highlights the diagnostic principles, classification updates, prognostic stratification systems, and novel treatments, which could inform future clinical trials and enhance the management of adult MDS patients, particularly for specific molecularly defined subgroups. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Venetoclax and hypomethylating agents in critically ill patients with newly diagnosed acute myeloid leukaemia.
- Author
-
Liang, Peiqi, Xie, Yan, Liu, Ziyi, Wang, Dong, Li, Qian, Lu, Yin, Xue, Shengli, Wang, Ying, Chen, Suning, Wu, Deipei, and Fu, Jianhong
- Subjects
- *
ACUTE myeloid leukemia , *APACHE (Disease classification system) , *VENETOCLAX , *CRITICALLY ill , *INTENSIVE care units - Abstract
Summary: Venetoclax (VEN) in combination with hypomethylating agents (HMAs) is considered the standard of treatment for individuals with newly diagnosed acute myeloid leukaemia (AML) who are ineligible for intensive chemotherapy. We conducted a retrospective analysis that encompassed 16 critically ill patients newly diagnosed with AML who were admitted to the intensive care unit (ICU) and received the VEN and HMA regimen. Among them, 13 were primary AML, and three were MDS‐transformed AML. The mean Acute Physiology and Chronic Health Evaluation II (APACHE II) score was 18.9, and the mean sepsis‐related organ failure assessment score (SOFA) was 6.2. The average length of the ICU stay was 27.3 days. The median duration of VEN administration was 16 days. After the first course of VEN + HMA, 12 cases (75%) achieved complete remission (CR) or CR with incomplete haematological recovery (CRi). Among the five patients harbouring TP53 mutations, the overall response rate (ORR) was 90%. All patients experienced grade 3–4 haematological adverse events (AEs). With a median follow‐up of 9.5 months (range: 0.5–23), the overall survival (OS) rate was 43.75%. TP53‐wild patients and CR state after the first course of VEN–HMA indicated better survival. The combination of VEN and HMA has demonstrated a significantly elevated therapeutic response rate in newly diagnosed AML patients with critical illness. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Hypomethylating agent monotherapy in core binding factor acute myeloid leukemia: a French multicentric retrospective study.
- Author
-
Gabellier, Ludovic, Peterlin, Pierre, Thepot, Sylvain, Hicheri, Yosr, Paul, Franciane, Gallego-Hernanz, Maria Pilar, Bertoli, Sarah, Turlure, Pascal, Pigneux, Arnaud, Guieze, Romain, Ochmann, Marlène, Malfuson, Jean-Valère, Cluzeau, Thomas, Thomas, Xavier, Tavernier, Emmanuelle, Jourdan, Eric, Bonnet, Sarah, Tudesq, Jean-Jacques, and Raffoux, Emmanuel
- Subjects
- *
ACUTE myeloid leukemia , *ERYTHROCYTES , *BLOOD platelets , *RETROSPECTIVE studies , *AZACITIDINE - Abstract
Very few data are available about hypomethylating agent (HMA) efficiency in core binding factor acute myeloid leukemias (CBF-AML). Our main objective was to evaluate the efficacy and safety of HMA in the specific subset of CBF-AML. Here, we report the results of a multicenter retrospective French study about efficacy of HMA monotherapy, used frontline or for R/R CBF-AML. Forty-nine patients were included, and received a median of 5 courses of azacitidine (n = 46) or decitabine (n = 3). ORR was 49% for the whole cohort with a median time to response of 112 days. After a median follow-up of 72.3 months, median OS for the total cohort was 10.6 months. In multivariate analysis, hematological relapse of CBF-AML at HMA initiation was significantly associated with a poorer OS (HR: 2.13; 95%CI: 1.04–4.36; p = 0.038). Responders had a significantly improved OS (1-year OS: 75%) compared to non-responders (1-year OS: 15.3%; p < 0.0001). Hematological improvement occurred for respectively 28%, 33% and 48% for patients who were red blood cell or platelet transfusion-dependent, or who experienced grade 3/4 neutropenia at HMA initiation. Adverse events were consistent with the known safety profile of HMA. Our study highlights that HMA is a well-tolerated therapeutic option with moderate clinical activity for R/R CBF-AML and for patients who cannot handle intensive chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Venetoclax-based therapy for relapsed or refractory acute myeloid leukemia: latest updates from the 2023 ASH annual meeting
- Author
-
Xubo Gong, Xin He, Lin Wang, Teng Yu, Weiwei Liu, Huiying Xu, Lan Jin, Xiang Li, Bin Zhang, Zhihua Tao, and Wenbin Qian
- Subjects
Venetoclax ,Acute myeloid leukemia ,Hypomethylating agents ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Patients with relapsed or refractory (R/R) acute myeloid leukemia (AML) often exhibit limited responses to traditional chemotherapy, resulting in poor prognosis. The combination of venetoclax (VEN) with hypomethylating agents has been established as the standard treatment for elderly or medically unfit AML patients unable to undergo intensive chemotherapy. Despite this, the availability of novel VEN-based therapies specifically tailored for those with R/R AML remains scarce. Here, we provide a comprehensive overview of the latest data presented at the 65th American Society of Hematology Annual Meeting, shedding light on the progress and efficacy of VEN-based therapies for R/R AML.
- Published
- 2024
- Full Text
- View/download PDF
28. Efficacy and safety of combination therapies vs monotherapy of hypomethylating agents in accelerated or blast phase of Philadelphia negative myeloproliferative neoplasms: a systematic review and meta-analysis
- Author
-
Jia Chen, Kefei Wang, Zhijian Xiao, and Zefeng Xu
- Subjects
Myeloproliferative neoplasms ,accelerated/blast phase ,hypomethylating agents ,ruxolitinib ,venetoclax ,Medicine - Abstract
AbstractBackground There is a lack of evidence regarding whether combination therapy of hypomethylating agents (HMAs) has better outcomes than HMA monotherapy in patients with Philadelphia chromosome-negative accelerated or blast phase myeloproliferative neoplasms (MPN-AP/BP).Materials and methods Pubmed, Embase, Web of Science and Cochrane library databases were searched for studies from inception of each database until 31 December 2021. Data extraction and synthesis were conducted following the PRISMA reporting guideline.Results It was found that HMAs plus venetoclax therapy yielded a higher CR/CRi rate than HMAs alone [36% vs 19%, p = .0204] and a higher CR rate than HMAs plus ruxolitinib [22% vs 8%, p = .0313]. HMAs plus ruxolitinib combination showed a higher ORR than HMA monotherapy [45% vs 30%, p = .0395], but there was no improvement in CR/CRi. The one-year and two-year OS rate for patients treated with HMAs plus venetoclx/ruxolitinib demonstrated a trend towards prolonged survival than HMAs alone [HMAs plus venetoclax: 24% vs 11%, p = .1295 and 12% vs 3%, p = .2357; HMAs plus ruxolitinib: 25% vs 11%, p = .0774 and 33% vs 3%, p = .051].Conclusion It was confirmed that HMA in combination with venetoclax is an effective and well-tolerated option in MPN-AP/BP patients in pre- as well as post-haematopoietic stem cell transplantation settings. HMA plus ruxolitinib therapy was revealed to be effective in patients with MPN-AP.Key MessagesCombination therapy with HMAs and venetoclax/ruxolitinib was associated with improved outcomes than HMAs alone in MPN-AP/BP patients.Further large-scale randomized controlled trials are needed to confirm regarding to the optimal treatment for this patient population.
- Published
- 2023
- Full Text
- View/download PDF
29. Azacitidine induced lung injury: report and contemporary discussion on diagnosis and management.
- Author
-
Alyamany, Ruah, Alnughmush, Ahmed, Almutlaq, Malak, Alyamany, Mohammed, and Alfayez, Mansour
- Subjects
LUNG injuries ,AZACITIDINE ,MYELODYSPLASTIC syndromes ,ACUTE myeloid leukemia ,STEM cell transplantation - Abstract
Azacitidine, a hypomethylating agent, has caused a paradigm shift in the outcomes of patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) who are not eligible for stem cell transplantation, particularly in combination with BCL2 and IDH inhibitors. Azacitidine and Azacitidine-based combinations have been widely considered a safe low-intensity therapy when compared to traditional conventional treatments. The development of lung toxicity from azacitidine is not a well-characterized adverse event. However, if it happens, it can be fatal, especially if not recognized and treated promptly. In this review, we aim to familiarize the reader with the presentation of azacitidine-induced lung injury, provide our suggested approach to management based on our experience and the current understanding of its mechanism, and review the literature of 20 case reports available on this topic. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Venetoclax-based therapy for relapsed or refractory acute myeloid leukemia: latest updates from the 2023 ASH annual meeting.
- Author
-
Gong, Xubo, He, Xin, Wang, Lin, Yu, Teng, Liu, Weiwei, Xu, Huiying, Jin, Lan, Li, Xiang, Zhang, Bin, Tao, Zhihua, and Qian, Wenbin
- Subjects
- *
ACUTE myeloid leukemia , *ANNUAL meetings , *VENETOCLAX - Abstract
Patients with relapsed or refractory (R/R) acute myeloid leukemia (AML) often exhibit limited responses to traditional chemotherapy, resulting in poor prognosis. The combination of venetoclax (VEN) with hypomethylating agents has been established as the standard treatment for elderly or medically unfit AML patients unable to undergo intensive chemotherapy. Despite this, the availability of novel VEN-based therapies specifically tailored for those with R/R AML remains scarce. Here, we provide a comprehensive overview of the latest data presented at the 65th American Society of Hematology Annual Meeting, shedding light on the progress and efficacy of VEN-based therapies for R/R AML. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Tumor lysis syndrome in patients with acute myeloid leukemia treated with venetoclax and hypomethylating agents with or without dose ramp-up.
- Author
-
Khouderchah, Christy J., Benitez, Lydia L., Marini, Bernard L., Fraga, Martina, Pettit, Kristen, Burke, Patrick W., Bixby, Dale L., and Perissinotti, Anthony J.
- Subjects
- *
TUMOR lysis syndrome , *ACUTE myeloid leukemia , *VENETOCLAX - Abstract
Venetoclax with hypomethylating agents (HMAs) is an important treatment for patients with acute myeloid leukemia (AML) who cannot tolerate intensive chemotherapy. However, there is limited data on the safety of venetoclax without a dose ramp-up in patients with AML. A retrospective cohort analysis of patients with AML treated with HMA/venetoclax (HMA/Ven) with or without a dose ramp-up, or HMA alone from 6/30/2014–8/22/2022 was conducted. The primary endpoint was the incidence of laboratory and/or clinical tumor lysis syndrome (TLS) by day 10. Of 225 patients, 111 patients received HMA alone or HMA/Ven with a dose ramp-up and 114 received HMA/Ven with no dose ramp-up. The incidence of TLS was similar between the control and no dose ramp-up groups, with rates of 5.4% and 5.3% respectively (p = 0.962). TLS incidence was comparable in patients with and without a dose ramp-up, suggesting that a dose ramp-up may not be mandatory in patients with AML. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Real-Life Multicenter Experience of Venetoclax in Combination with Hypomethylating Agents in Previously Untreated Adult Patients with Acute Myeloid Leukemia in Greece.
- Author
-
Chatzilygeroudi, Theodora, Darmani, Ismini, El Gkotmi, Natali, Vryttia, Pinelopi, Douna, Stavroula, Bouchla, Anthi, Labropoulou, Vasiliki, Kotsopoulou, Maria, Symeonidis, Argiris, Pagoni, Maria, Pappa, Vasiliki, and Papageorgiou, Sotirios G.
- Subjects
- *
ACUTE myeloid leukemia , *VENETOCLAX , *MYCOSES , *ADULTS , *SURVIVAL rate - Abstract
Background: The landscape of first-line treatment for acute myeloid leukemia (AML) patients ineligible for intensive chemotherapy has changed remarkably after venetoclax approval. Accumulating real-world data further apprises us with more knowledgeable use. To assess the efficacy and safety challenges in the real-life setting of the combination of hypomethylated agent (HMA) and venetoclax, we conducted a multi-center retrospective study. Methods: Forty adult AML patients treated with the combination of HMA and venetoclax as a first-line treatment after full approval (2020) were included. To confirm VIALE-A results, this group was compared to a historical cohort of 17 chemotherapy-ineligible AML patients treated with HMA monotherapy before 2020. Results: The combination of HMA-venetoclax achieved a composite complete response rate of 86.8% (p < 0.001), median overall survival, and event-free survival of 33.8 and 19.7 months, respectively, in a median follow-up of 17.8 months (pos < 0.001, HR = 0.276, CI: 0.132–0.575, pEFS = 0.004, HR = 0.367, CI: 0.174–0.773). High rates of neutropenia (90%) and consequent infection rates (57.5%) were noted. Only 55% of our patients received antifungal prophylaxis, as its use remains controversial, and invasive fungal infections were presented in 7.5%. Conclusions: Evidently, venetoclax-HMA yields high response rates and profound survival benefits in real life and has changed our approach to alternative chemotherapy options. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Clinical and genetic characteristics predict outcomes of acute myeloid leukemia patients with FLT3 mutations receiving venetoclax‐based therapy.
- Author
-
Weng, Guangyang, Huang, Jingya, An, Na, Zhang, Yu, Yu, Guopan, Sun, Zhiqiang, Lin, Dongjun, Deng, Lan, Liang, Xinquan, Xiao, Jie, Zhang, Hongyu, Guo, Ziwen, He, Xin, Jin, Hua, Liu, Qifa, and Du, Xin
- Subjects
- *
ACUTE myeloid leukemia , *PROPORTIONAL hazards models - Abstract
Background: Acute myeloid leukemia (AML) is a heterogeneous disease, and its heterogeneity is associated with treatment response. Despite the demonstrated success of venetoclax (VEN)‐based therapy for AML, the effect of FLT3 mutations on the efficacy of the therapy is poorly understood. We aimed to compare the efficacy of VEN‐based therapy between FLT3‐mutated (FLT3mut) and FLT3 wild‐type (FLT3wt) patients and identify the predictors of efficacy in FLT3mut patients. Methods: A total of 266 AML patients (127 newly diagnosed [ND] and 139 refractory/relapsed [R/R]) receiving VEN‐based regimens were enrolled in this study. A retrospective analysis was performed, and the treatment responses and overall survival (OS) of FLT3mut and FLT3wt patients were compared. Logistic regression and Cox proportional hazards model were applied to examine the clinical and genetic predictors of outcomes. Results: With a median of two cycles of VEN‐based therapy, for the ND AML cohort, the FLT3mut group had a comparable composite complete remission (CRc) rate with the FLT3wt group (79.3% vs. 61.2%, p = 0.072). For the R/R AML cohort, the FLT3mut group exhibited a lower CRc rate than the FLT3wt group. With a median follow‐up of 8.6 months (95% confidence interval [CI], 8.0–10), the median OS observed in the FLT3mut and FLT3wt groups for both cohorts were close (14.0 vs. 19.9 months, p = 0.356; 10.0 vs. 11.9 months, p = 0.680). For the ND AML cohort, in FLT3mut patients, MRD‐positive and RNA‐splicing mutation predicted inferior survival (hazard ratio [HR], 10.3; 95% CI: 2.0–53.8; p = 0.006; HR 11.3; 95% CI: 1.2–109.3; p = 0.036, respectively). For the R/R AML cohort, in FLT3mut patients, adverse ELN risk was associated with an inferior response (odds ratio [OR], 0.2; 95% CI: 0.1–0.8; p = 0.025), whereas NPM1 co‐mutation was associated with a superior response (57.1%; OR, 6.7; 95% CI: 1.5–30.1; p = 0.014). CR/CRi predicted a better survival (HR 0.2; 95% CI: 0.1–0.8; p = 0.029), while DNMT3A mutation predicted an inferior survival (HR, 4.6; 95% CI: 1.4–14.9; p = 0.011). Conclusions: FLT3 mutations may influence response to VEN‐based therapy in R/R AML patients but not in ND AML patients. Furthermore, clinical and genetic characteristics could predict outcomes of FLT3mut patients receiving VEN‐based therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Venetoclax plus hypomethylating agents in DDX41‐mutated acute myeloid leukaemia and myelodysplastic syndrome: Mayo Clinic series on 12 patients.
- Author
-
Nanaa, Ahmad, He, Rong, Foran, James M., Badar, Talha, Gangat, Naseema, Pardanani, Animesh, Hogan, William J., Litzow, Mark R., Patnaik, Mrinal, Al‐Kali, Aref, and Alkhateeb, Hassan B.
- Subjects
- *
ACUTE myeloid leukemia , *MYELODYSPLASTIC syndromes , *VENETOCLAX , *THERAPEUTICS - Abstract
Summary: Venetoclax (VEN) is an FDA‐approved selective inhibitor of B‐cell leukaemia/lymphoma‐2 (BCL‐2), used for treating elderly or unfit acute myeloid leukaemia (AML) patients unable to undergo intensive chemotherapy. Combining VEN with hypomethylating agents (HMAs) has shown impressive response rates in high‐risk myelodysplastic syndromes (MDS) and relapsed/refractory AML. However, the efficacy of VEN and HMAs in treating DDX41‐mutated (mDDX41) MDS/AML patients remains uncertain. Despite the favourable prognostic nature of mDDX41 MDS/AML patients, there is a lack of clinical experience regarding their response to different treatment regimens, leading to an unknown optimal therapeutic approach. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
35. Venetoclax and Hypomethylating Agent Combination in Myeloid Malignancies: Mechanisms of Synergy and Challenges of Resistance.
- Author
-
Mishra, Rahul, Zokaei Nikoo, Maedeh, Veeraballi, Sindhusha, and Singh, Abhay
- Subjects
- *
VENETOCLAX , *ACUTE myeloid leukemia , *BCL genes , *OXIDATIVE phosphorylation , *TREATMENT effectiveness , *STEM cells - Abstract
There has been a widespread adoption of hypomethylating agents (HMA: 5-Azacytidine (5-Aza)/decitabine) and venetoclax (Ven) for the treatment of acute myeloid leukemia (AML); however, the mechanisms behind the combination's synergy are poorly understood. Monotherapy often encounters resistance, leading to suboptimal outcomes; however, the combination of HMA and Ven has demonstrated substantial improvements in treatment responses. This study elucidates multiple synergistic pathways contributing to this enhanced therapeutic effect. Key mechanisms include HMA-mediated downregulation of anti-apoptotic proteins, notably MCL-1, and the priming of cells for Ven through the induction of genes encoding pro-apoptotic proteins such as Noxa. Moreover, Ven induces sensitization to HMA, induces overcoming resistance by inhibiting the DHODH enzyme, and disrupts antioxidant pathways (Nrf2) induced by HMA. The combination further disrupts oxidative phosphorylation in leukemia stem cells, amplifying the therapeutic impact. Remarkably, clinical studies have revealed a favorable response, particularly in patients harboring specific mutations, such as IDH1/2, NPM1, CEBPA, or ASXL1. This prompts future studies to explore the nuanced underpinnings of these synergistic mechanisms in AML patients with these molecular signatures. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Venetoclax combined with hypomethylating agents and the CAG regimen in relapsed/refractory AML: a single-center clinical trial.
- Author
-
Yifan Liu, Yanfen Li, Ran Zhang, Zhangyu Yu, and Yu Jing
- Subjects
HEMATOPOIETIC stem cell transplantation ,VENETOCLAX ,ACUTE myeloid leukemia ,PATIENT experience ,CLINICAL trials - Abstract
Objective: This study aimed to evaluate the efficacy and safety of venetoclax in combination with hypomethylating agents and CAG (VEN-DCAG) regimens in patients with relapsed/refractory acute myeloid leukemia (R/R AML). Methods: The treatment response was analyzed by retrospective methods in R/R AML patients treated with the VEN-DCAG regimen at our institution. This included, but was not limited to, CR/CRi (complete remission/complete remission with incomplete hematologic recovery) rate, measurable residual disease (MRD) negative rate, and overall survival (OS). Results: 20 patients with R/R AML were recruited, with a median age of 40 years (10-70), 11 of whomwere male (55%), and a median follow-up of 10.4 months (0.7-21.8). The overall response rate (ORR) after receiving 1 course of VEN-DCAG was 90% (18/20), with 17 (85%) CR/CRi (10 MRD-CR), 1 (5%) PR, and 2 (10%) NR. Subsequently, 12 patients (7 MRD-CR, 4 MRD+CR, 1 NR) were treated with the VEN-DCAG regimen, and 3 MRD+CR patients turned negative, and 13 patients finally achieved MRD-CR. Among them, 7 patients were in the relapse group, all achieving CR/CRi (6 MRD-CR), and 13 patients in the refractory group, with 10 CR/CRi (7 MRD-CR). The ORR for patients in the relapse and refractory groups was 100% (7/7) and 84.6% (11/13), respectively. Further, all patients experienced adverse events (AEs) of varying degrees of severity, with hematologic AEs primarily consisting of myelosuppression, while non-hematologic AEs were more common in the form of fever, gastrointestinal distress, and infections. 11 patients were followed upwith bridging allogeneic hematopoietic stem cell transplantation (allo-HSCT) therapy. At the last follow-up, 11 patients (7MRD-CR, 4MRD+CR) who received allo-HSCT, 1 (MRD+CR) died, and 9 patients (6 MRD-CR, 1 PR, 2 NR) who did not receive allo-HSCT, 5 (2 MRD-CR, 1 PR, 2 NR) died as well. Conclusion: The VEN-DCAG regimen may be an effective treatment option for R/R AML patients, with high ORR and MRD negative remission rates in both the relapsed and refractory groups. It is recommend that patients should be bridged to allo-HSCT as soon as possible after induction to CR by the VEN-DCAG regimen, which can lead to a significant long-term survival benefit. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
37. Real-world results of venetoclax combined with hypomethylating agents in young adults with relapsed/refractory acute myeloid leukemia.
- Author
-
Xu, Xuezhu, Liu, Rui, He, Aili, and Wang, Fangxia
- Subjects
- *
ACUTE myeloid leukemia , *YOUNG adults , *HEMATOPOIETIC stem cell transplantation , *VENETOCLAX - Abstract
Young adults with acute myeloid leukemia (AML) often fail to achieve permanent complete remission (CR) and frequently relapse, indicating an urgent need to explore effective salvage therapies. Recent advances in AML treatment have been attributed to the combination of the B-cell lymphoma 2 (Bcl-2) inhibitor venetoclax (VEN) with hypomethylating agents (HMAs); however, the use of this combination in young adults with relapsed or refractory (R/R) AML has not been reported. We retrospectively examined 31 young patients with R/R AML treated with VEN plus an HMA. We evaluated the demographic data, cytogenetic characteristics, AML types, response rates, and transplantation-related data for the patients in our cohort. The combination of VEN + HMA yielded a CR rate of 48.4%. The most prominent hematologic adverse event was neutropenia, which occurred in all patients, with 90.3% of cases being grade ≥3. Non-hematologic toxicities were relatively mild and infrequent, with an incidence of 45.2%. More than half of the patients with sustained CR had received an allogeneic hematopoietic stem cell transplantation (allo-HSCT), of whom two died of transplant-related complications. Our results showed that the combination of VEN + HMA appeared to be a highly effective and well-tolerated salvage therapy option for young patients with R/R AML, enabling more young patients to proceed to potentially curative allo-HSCT. However, additional, well-designed studies with larger numbers of patients are required to confirm the advantages of VEN + HMA in this population. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. Combination of venetoclax with BCR-ABL tyrosine kinase inhibitor as a therapeutic strategy for Philadelphia chromosome-positive leukemias.
- Author
-
Zou, Jing-Ying, Huang, Si-Man, Zhoub, Hai-Xia, Xu, Ming-Zhu, Sun, Ai-Ning, Wu, De-Pei, Xue, Sheng-Li, and Zhang, Tong-Tong
- Subjects
- *
PROTEIN-tyrosine kinase inhibitors , *LEUKEMIA , *VENETOCLAX , *CHRONIC myeloid leukemia , *ACUTE myeloid leukemia , *AZACITIDINE - Abstract
Venetoclax has shown synergism with BCR-ABL1 tyrosine kinase inhibitors (TKIs) in preclinical studies for patients with Philadelphia chromosome-positive (Ph+) leukemias. This combination may suggest a novel treatment strategy for Ph + leukemias. We conducted a retrospective study to summarize the activity of combining venetoclax and BCR-ABL1 TKI-based therapies in Ph + leukemias. A total of 18 patients with Ph + leukemias were enrolled in this study. At the time of venetoclax and TKI-based therapy, 5 patients were initially diagnosed, with Ph + acute myeloid leukemia (AML) (n = 1) and mixed phenotype acute leukemia (MPAL) (n = 4), 7 patients had chronic myeloid leukemia at blastic phase (CML-BP), and the remaining 6 patients had relapsed or refractory to prior therapy. The overall response rate (ORR) was 88.9% (9 CR, 2 CRi, 4 MLFS, 1 PR), and a major molecular response (MMR) (or better) was achieved in 7 (38.8%) of all patients. With a median follow-up of 7.0 months (range, 2.3-15.6), 15 (83.3%) were in continuous CR at the time of this analysis, with a 1-year OS of 85.6%, 1-year LFS of 76.7%, and 1-year CIR of 22.4%. Moreover, 10 of 18 patients were treated with venetoclax, TKI and hypomethylating agent (HMA) regimens, which also associated with a high ORR rate (6 CR, 1 CRi, 3 MLFS), and can be used for induction or salvage therapy. Venetoclax and TKI-based combination regimens may be a feasible approach for Ph + leukemias, and prospective studies are needed to properly assess the safety, tolerability and efficacy of this regimen. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. Hypomethylating agents plus modified priming regimens compared with venetoclax-based regimens based on molecular characteristics for newly diagnosed patients with acute myeloid leukemia: a multi-center cohort study.
- Author
-
Weng, Guangyang, Huang, Jingya, He, Xin, Xue, Tingting, Yang, Linlin, Zhang, Yu, Yu, Guopan, Sun, Zhiqiang, Lin, Dongjun, Deng, Lan, Liang, Xinquan, Xiao, Jie, Zhang, Hongyu, Guo, Ziwen, Jin, Hua, Liu, Qifa, and Du, Xin
- Subjects
- *
ACUTE myeloid leukemia , *COHORT analysis , *OLDER patients , *VENETOCLAX - Abstract
Venetoclax (VEN)-based regimens are the standard of care for elderly or unfit patients with newly diagnosed (ND) acute myeloid leukemia (AML). Some single-arm studies have implied that hypomethylating agents (HMAs) plus priming regimens may potentially provide an alternative therapeutic approach, owing to encouraging efficacy seen. However, no comparative data exists yet regarding these two treatment approaches. In this retrospective multi-center cohort study, we enrolled 294 ND AML patients, allocating 167 to the HMA + priming group and 127 to the VEN-based group. Treatment response and overall survival (OS) were compared between groups. Molecular subgroup analyses were also conducted. With a median of two cycles for HMA + priming group, the overall response (ORR) was 65.3%, including 55.1% complete remission (CR), 9.6% CR with incomplete hematologic recovery (CRi) and 0.6% morphologic leukemia-free state (MLFS). With a median of two cycles for VEN-based group, the ORR was 70.9%, including 46.5% CR, 18.9% CRi, and 5.5% MLFS. Response differences (ORR or CR/CRi) between groups were not significant (p > 0.05). With a median follow-up of 10.1 months, median OSs were similar between groups (20.9 vs 16.3 months, p = 0.41). However, VEN regimens demonstrated superior CR/CRi for patients with mutations in FLT3, IDH1/2, and NPM1 compared to HMA + priming (80.0% vs 35.0%, p = 0.01; 90.9% vs 65.5%, p = 0.02; 90.9% and 65.5%, p = 0.02, respectively). In conclusion, HMAs plus modified priming regimens might be a potential alternative therapeutic approach for patients with ND AML, but VEN-based regimens presented predominance in specific molecular subgroups. Molecular characteristics contribute to guiding choice of treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
40. Efficacy and safety of combination therapies vs monotherapy of hypomethylating agents in accelerated or blast phase of Philadelphia negative myeloproliferative neoplasms: a systematic review and meta-analysis.
- Author
-
Chen, Jia, Wang, Kefei, Xiao, Zhijian, and Xu, Zefeng
- Subjects
MYELOPROLIFERATIVE neoplasms ,STEM cell transplantation ,VENETOCLAX ,RUXOLITINIB ,DATA extraction - Abstract
There is a lack of evidence regarding whether combination therapy of hypomethylating agents (HMAs) has better outcomes than HMA monotherapy in patients with Philadelphia chromosome-negative accelerated or blast phase myeloproliferative neoplasms (MPN-AP/BP). Pubmed, Embase, Web of Science and Cochrane library databases were searched for studies from inception of each database until 31 December 2021. Data extraction and synthesis were conducted following the PRISMA reporting guideline. It was found that HMAs plus venetoclax therapy yielded a higher CR/CRi rate than HMAs alone [36% vs 19%, p =.0204] and a higher CR rate than HMAs plus ruxolitinib [22% vs 8%, p =.0313]. HMAs plus ruxolitinib combination showed a higher ORR than HMA monotherapy [45% vs 30%, p =.0395], but there was no improvement in CR/CRi. The one-year and two-year OS rate for patients treated with HMAs plus venetoclx/ruxolitinib demonstrated a trend towards prolonged survival than HMAs alone [HMAs plus venetoclax: 24% vs 11%, p =.1295 and 12% vs 3%, p =.2357; HMAs plus ruxolitinib: 25% vs 11%, p =.0774 and 33% vs 3%, p =.051]. It was confirmed that HMA in combination with venetoclax is an effective and well-tolerated option in MPN-AP/BP patients in pre- as well as post-haematopoietic stem cell transplantation settings. HMA plus ruxolitinib therapy was revealed to be effective in patients with MPN-AP. Combination therapy with HMAs and venetoclax/ruxolitinib was associated with improved outcomes than HMAs alone in MPN-AP/BP patients. Further large-scale randomized controlled trials are needed to confirm regarding to the optimal treatment for this patient population. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. Ex vivo characterization of acute myeloid leukemia patients undergoing hypomethylating agents and venetoclax regimen reveals a venetoclax-specific effect on non-suppressive regulatory T cells and bona fide PD-1+TIM3+ exhausted CD8+ T cells
- Author
-
Giulia Corradi, Dorian Forte, Gianluca Cristiano, Andrea Polimeno, Marilena Ciciarello, Valentina Salvestrini, Lorenza Bandini, Valentina Robustelli, Emanuela Ottaviani, Michele Cavo, Darina Ocadlikova, and Antonio Curti
- Subjects
acute myeloid leukemia ,immune system ,venetoclax ,hypomethylating agents ,immune checkpoints receptors ,immune checkpoint inhibitors (ICIs) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Acute myeloid leukemia (AML) is an aggressive heterogeneous disease characterized by several alterations of the immune system prompting disease progression and treatment response. The therapies available for AML can affect lymphocyte function, limiting the efficacy of immunotherapy while hindering leukemia-specific immune reactions. Recently, the treatment based on Venetoclax (VEN), a specific B-cell lymphoma 2 (BCL-2) inhibitor, in combination with hypomethylating agents (HMAs) or low-dose cytarabine, has emerged as a promising clinical strategy in AML. To better understand the immunological effect of VEN treatment, we characterized the phenotype and immune checkpoint (IC) receptors’ expression on CD4+ and CD8+ T cells from AML patients after the first and second cycle of HMA in combination with VEN. HMA and VEN treatment significantly increased the percentage of naïve CD8+ T cells and TIM-3+ CD4+ and CD8+ T cells and reduced cytokine-secreting non-suppressive T regulatory cells (Tregs). Of note, a comparison between AML patients treated with HMA only and HMA in combination with VEN revealed the specific contribution of VEN in modulating the immune cell repertoire. Indeed, the reduction of cytokine-secreting non-suppressive Tregs, the increased TIM-3 expression on CD8+ T cells, and the reduced co-expression of PD-1 and TIM-3 on both CD4+ and CD8+ T cells are all VEN-specific. Collectively, our study shed light on immune modulation induced by VEN treatment, providing the rationale for a novel therapeutic combination of VEN and IC inhibitors in AML patients.
- Published
- 2024
- Full Text
- View/download PDF
42. Efficacy and safety of venetoclax plus hypomethylating agents in relapsed/refractory acute myeloid leukemia: a multicenter real-life experience
- Author
-
Francesco Angotzi, Federica Lessi, Matteo Leoncin, Carla Filì, Mauro Endri, Albana Lico, Andrea Visentin, Stefano Pravato, Anna Candoni, Livio Trentin, and Carmela Gurrieri
- Subjects
acute myeloid leukemia ,venetoclax ,hypomethylating agents ,azacitidine ,decitabine ,relapsed/refractory acute myeloid leukemia ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Venetoclax (VEN) has been shown to play a synergistic effect in combination with hypomethylating agents (HMAs) in the frontline treatment of acute myeloid leukemia (AML). However, the potential role of this therapy in the relapsed/refractory (R/R) AML setting, still needs to be further unveiled. The aim of the current study was to retrospectively outline the safety profile, response and survival outcomes of R/R AML patients treated with VEN in association with HMAs. Clinical, biological, and molecular data were collected from 57 patients with R/R AML treated with VEN combined with azacitidine or decitabine between 2018 and 2023. The median age of patients was 63 years, 38 (66.7%) received treatment for relapsed disease while 19 (33.3%) for refractory disease, 5 (8.7%) were treated for molecular relapse. A consistent proportion of the cohort was represented by patients with unfavorable prognostic factors such as complex karyotype (36.8%), secondary AML (29.8%), previous exposure to HMAs (38.6%), and relapse after allogeneic stem cell transplant (22.8%). A total of 14 patients achieved CR (24.6%), 3 (5.3%) CRi, 3 (5.3%) MLFS, and 3 (5.3%) PR, accounting for an ORR of 40.4%. The CR/CRi rate was higher in the group treated with azacitidine than in the group treated with decitabine (37.8% vs. 15%). The median OS was 8.2 months, reaching 20.1 months among responding patients. VEN-HMAs treatment allowed to bridge to allogeneic stem cell transplantation 11 (23.9%) of eligible patients, for which a median OS of 19.8 months was shown. On multivariate analysis, ECOG performance status ≥2, complex karyotype and not proceeding to allogeneic stem cell transplantation after therapy with VEN-HMAs were the factors independently associated with shorter OS. Patients treated with the azacitidine rather than the decitabine containing regimen generally displayed a trend toward superior outcomes. The major toxicities were prolonged neutropenia and infections. In conclusion, this study showed how VEN-HMAs could represent an effective salvage therapy in patients with R/R AML, even among some of those patients harboring dismal prognostic features, with a good toxicity profile. Further prospective studies are thus warranted.
- Published
- 2024
- Full Text
- View/download PDF
43. Editorial: Hematopoietic stem cell transplantation: back to the future
- Author
-
Erden Atilla
- Subjects
hematopoietic stem cell transplantation ,acute graft vs. host disease ,gastrointestinal GVHD ,GVHD prophylaxis ,hypomethylating agents ,XMEN syndrome ,Medicine (General) ,R5-920 - Published
- 2024
- Full Text
- View/download PDF
44. Post‐remission cytopenia management in patients with AML treated with venetoclax in combination with hypomethylating agents: Pre‐ versus post‐VIALE‐A real‐world experience from a predominantly US community setting
- Author
-
Pankit Vachhani, Esprit Ma, Tao Xu, Melissa Montez, Sarah Worth, Archibong Yellow‐Duke, Wei‐Han Cheng, Michael E. Werner, Jonathan Abbas, and William Donnellan
- Subjects
acute myeloid leukemia ,bone marrow assessment ,hypomethylating agents ,real‐world ,schedule modifications ,survival outcomes ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background This retrospective cohort study used an electronic health record‐derived, de‐identified, US patient‐level database to better understand the real‐world treatment experience, in a predominantly community setting (80.3% of patients), of venetoclax+hypomethylating agents (HMAs) in routine clinical care, pre‐ and post‐VIALE‐A, to determine whether the post‐remission cytopenia management insight from VIALE‐A was reflected in real‐world clinical practice. Methods Patients with newly diagnosed acute myeloid leukemia (AML; N = 498), who initiated venetoclax+HMA ≤30 days from AML diagnosis from June 1, 2018, to March 31, 2021, were stratified into pre‐(n = 330) and post‐(n = 168) VIALE‐A cohorts. Results More patients in the post‐(61%) versus pre‐(45%) VIALE‐A cohort had their first biopsy by 28 ± 14 days post‐treatment initiation. Patients underwent bone marrow (BM) assessment earlier in the post‐ versus pre‐VIALE‐A cohort, and first identification of response was also earlier (2.5 vs 5.1 months, respectively). More venetoclax schedule modifications post‐remission occurred among post‐(82.1%) versus pre‐(73.8%) VIALE‐A responders; the most common reason for modification was treatment toxicities, specifically cytopenia. Treatment survival outcomes were comparable with or without venetoclax schedule modifications. Conclusions Findings suggest that venetoclax schedule modifications can be used to manage cytopenia events without adversely affecting outcomes. Opportunities remain to improve earlier BM assessment to determine venetoclax schedule modifications, providing the best chance for optimal treatment outcomes.
- Published
- 2023
- Full Text
- View/download PDF
45. Decitabine/Cedazuridine in the Management of Myelodysplastic Syndrome and Chronic Myelomonocytic Leukemia in Canada
- Author
-
John Paul Yun, Philip Q. Ding, Aastha Dolley, and Winson Y. Cheung
- Subjects
myelodysplastic syndrome ,chronic myelomonocytic leukemia ,real-world evidence ,hypomethylating agents ,treatment patterns ,patient support programs ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The management of myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) is limited and remains an unmet need. Decitabine/cedazuridine (DEC-C, ASTX727) is Canada’s first and only approved oral hypomethylating agent for MDS and CMML. We characterized the real-world use of DEC-C through a Canadian compassionate use program. Demographic and clinical data from 769 patients enrolled in Taiho Pharma Canada’s Patient Support Program were collected and analyzed. These patients represent a collection period from 10 November 2020 to 31 August 2022 with a median age of 76 years. Among 651 patients who started DEC-C, the median treatment duration was 4.2 cycles. The median overall and progression-free survival were 21.6 and 10.7 months, respectively. Among 427 patients who discontinued treatment, the majority (69.5%) stopped due to death (n = 164) or disease progression (n = 133). Multivariable cox regression showed that age, province of residence, blast counts, antibiotic prophylaxis, and number of dose reductions and delays were not significantly associated with overall and progression-free survival. DEC-C is a promising alternative to parenteral hypomethylating agent therapy, and it likely addresses an important unmet need for effective and convenient therapies in this setting.
- Published
- 2023
- Full Text
- View/download PDF
46. Efficacy and safety of venetoclax combined with hypomethylating agents for relapse of acute myeloid leukemia and myelodysplastic syndrome post allogeneic hematopoietic stem cell transplantation: a systematic review and meta-analysis
- Author
-
Yufeng Du, Chunhong Li, Zhijia Zhao, Yikun Liu, Chengtao Zhang, and Jinsong Yan
- Subjects
Acute myeloid leukemia ,Myelodysplastic syndrome ,Venetoclax ,Hypomethylating agents ,Allogeneic hematopoietic cell transplantation ,Relapse ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Currently, there is no standard treatment for managing relapse in patients with acute myeloid leukemia and myelodysplastic syndrome (AML/MDS) after allogeneic hematopoietic cell transplantation. Venetoclax-based therapies have been increasingly used for treating post-transplantation relapse of AML. The aim of this systematic review and meta-analysis was to evaluate the efficacy and adverse events of Venetoclax combined with hypomethylating agents (HMAs) for AML/MDS relapse post-transplantation. Methods We searched PubMed, Web of Science, Excerpta Medica Database, Cochrane Library, and Clinical. gov for eligible studies from the inception to February 2022. The Methodological Index for Non-Randomized Studies was used to evaluate the quality of the included literatures. The inverse variance method calculated the pooled proportion and 95% confidence interval (CI). Results This meta-analysis included 10 studies involving a total of 243 patients. The pooled complete response and complete response with incomplete blood count recovery rate of Venetoclax combined with HMAs for post-transplantation relapse in AML/MDS was 32% (95% CI, 26-39%, I2 = 0%), with an overall response rate of 48% (95% CI, 39-56%, I2 = 37%). The 6-month survival rate was 42% (95% CI, 29-55%, I2 = 62%) and the 1-year survival rate was 23% (95% CI, 11-38%, I2 = 78%). Conclusion This study demonstrated a moderate benefit of Venetoclax in combination with HMAs for patients with relapsed AML/MDS post-transplantation (including those who have received prior HMAs therapy), and may become one of treatment options in the future. Large-scale prospective studies are needed to confirm the potential benefit from venetoclax combined with HMAs.
- Published
- 2023
- Full Text
- View/download PDF
47. Venetoclax-based therapy for relapsed or refractory acute myeloid leukemia: latest updates from the 2022 ASH annual meeting
- Author
-
Xubo Gong, Yi Zhang, Xin He, Milad Moloudizargari, Teng Yu, Lin Wang, Weiwei Liu, Lan Jin, Huiying Xu, Yang Xu, Zhihua Tao, and Wenbin Qian
- Subjects
Venetoclax ,Acute myeloid leukemia ,Hypomethylating agents ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Venetoclax (VEN), the first selective Bcl-2 inhibitor, has shown efficacy and safety both as monotherapy and in combination with other agents in the treatment of newly diagnosed acute myeloid leukemia (AML), while its role in relapsed or refractory (R/R) disease is not well defined. Here, we reviewed the latest advances of VEN-based therapy for R/R AML from the 2022 American Society of Hematology (ASH) Annual Meeting, including some novel and encouraging regimes, such as VCA, VAH, and HAM regimes, etc. Further research is still needed to fully understand the optimal use of these agents in R/R AML treatment.
- Published
- 2023
- Full Text
- View/download PDF
48. Azacitidine induced lung injury: report and contemporary discussion on diagnosis and management
- Author
-
Ruah Alyamany, Ahmed Alnughmush, Malak Almutlaq, Mohammed Alyamany, and Mansour Alfayez
- Subjects
azacitidine induced lung injury myelodysplastic syndrome ,acute myeloid leukemia ,azaciditine ,hypomethylating agents ,acute lung injury ,pneumonitis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Azacitidine, a hypomethylating agent, has caused a paradigm shift in the outcomes of patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) who are not eligible for stem cell transplantation, particularly in combination with BCL2 and IDH inhibitors. Azacitidine and Azacitidine-based combinations have been widely considered a safe low-intensity therapy when compared to traditional conventional treatments. The development of lung toxicity from azacitidine is not a well-characterized adverse event. However, if it happens, it can be fatal, especially if not recognized and treated promptly. In this review, we aim to familiarize the reader with the presentation of azacitidine-induced lung injury, provide our suggested approach to management based on our experience and the current understanding of its mechanism, and review the literature of 20 case reports available on this topic.
- Published
- 2024
- Full Text
- View/download PDF
49. Real-world treatments and clinical outcomes in unfit AML patients receiving first-line treatment or best supportive care in Italy (CURRENT study)
- Author
-
Maria Paola Martelli, Nicola Di Renzo, Antonio Curti, Nicola Stefano Fracchiolla, Luca Maurillo, Morena Caira, Paola Finsinger, Giuliana Gualberti, Felicetto Ferrara, and Attilio Olivieri
- Subjects
Acute myeloid leukemia ,Italy ,Treatment pattern ,Elderly patients ,Hypomethylating agents ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Real-world data on treatment patterns and outcomes of patients with acute myeloid leukemia unfit for intensive chemotherapy are lacking before the advent of precision medicine in this setting.Herein, we present the Italian sub-analysis of the CURRENT study in AML patients unfit for first line intensive chemotherapy, evaluating patients’ outcomes between 2015 and 2018.Among 74 evaluable patients, 62 received systemic treatments (most used therapy was hypomethylating agents), while 12 best supportive care.Key results include both efficacy and safety data, as well as HCRU and treatment patterns. In first-line therapy cohort median OS was 13.4 months vs. 2.7 months for BSC.
- Published
- 2024
- Full Text
- View/download PDF
50. Long-term hematologic response after azacitidine treatment in a lower-risk myelodysplastic syndrome patient: A case report
- Author
-
Konstantinos Loukidis and Marcel Tschopp
- Subjects
Myelodysplastic syndrome ,Lower-risk MDS ,Hypomethylating agents ,Azacitidine ,Hematological response ,Case report ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
We report results of a 65-year-old patient with lower-risk myelodysplastic syndrome and multilineage dysplasia treated with hypomethylating agents. After failure of erythropoietin and thalidomide, the patient received azacitidine and achieved hematological remission for 95 months. In 2016, the treatment was switched to decitabine with promising results. These data showed that azacitidine used as a third-line treatment resulted in an exceptionally long-lasting positive hematological response after standard first- and second-line therapies had failed. Additionally, the patient experienced a good quality of life with no complications related to profound cytopenia, and continues to do so at the time of this report's preparation.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.