1
|
Franco S, Khan T, Dinner S, Karmali R, Melody M. Dosing of 7 + 3 induction chemotherapy in a patient with acute myeloid leukemia (AML) and morbid obesity. J Oncol Pharm Pract 2024; 30:945-949. [PMID: 38509800 DOI: 10.1177/10781552241240444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Traditional chemotherapy dosing is based on body surface area (BSA) using standard formulas, which can pose challenges in dosing patients at body weight extremes. Studies suggest that chemotherapy dosing according to actual body weight does not increase toxicity in obese patients and current guidelines recommend full weight-based dosing of chemotherapy regardless of body mass index (BMI). However, the dosing of anthracyclines in obese patients can be challenging given limitations in maximum cumulative dosage, particularly in those at very extreme BMI. In this case, we highlight the difficulties of dosing anthracycline-based induction chemotherapy in a patient with newly diagnosed acute myeloid leukemia (AML) and BMI >90 kg/m2. CASE REPORT A 40-year-old female with morbid obesity is diagnosed with AML (nucleophosmin 1 (NPMI) and isocitrate dehydrogenase-2 mutated, FMS-like tyrosine kinase 3-Internal tandem duplication negative). MANAGEMENT AND OUTCOME The patient was initiated on induction therapy with 7 + 3 with dose capping of BSA at 2.75 m2 (cytarabine 200 mg/m2 continuous infusion over 24 h for 7 days, plus daunorubicin 60 mg/m2 slow intravenous push for 3 days), followed by two cycles of high-dose cytarabine consolidation therapy using actual BSA. The patient achieved morphologic complete remission; however, measurable residual disease testing for NPM1 remained positive after induction therapy. DISCUSSION This case suggests that dose capping of anthracyclines in the treatment of newly diagnosed AML may be an effective and safe treatment alternative in those with extreme BMI elevations beyond what has been studied in the literature. Given the increasing incidence of morbid obesity, further studies are needed to confirm appropriate dosing of anthracycline-based regimens at upper BMI extremes (>60 kg/m2).
Collapse
Affiliation(s)
- Stephanie Franco
- Department of Internal Medicine, Northwestern Medicine, Chicago, IL, USA
| | - Talha Khan
- Robert H. Lurie Comprehensive Cancer Center, Department of Pharmacy, Chicago, IL, USA
| | - Shira Dinner
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Reem Karmali
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Megan Melody
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
2
|
Pötgens SA, Lecop S, Havelange V, Li F, Neyrinck AM, Neveux N, Maertens J, Walter J, Schoemans H, Delzenne NM, Bindels LB. Gut microbiota alterations induced by intensive chemotherapy in acute myeloid leukaemia patients are associated with gut barrier dysfunction and body weight loss. Clin Nutr 2023; 42:2214-2228. [PMID: 37806074 DOI: 10.1016/j.clnu.2023.09.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/01/2023] [Accepted: 09/21/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND & AIMS Acute myeloid leukaemia (AML) chemotherapy has been reported to impact gut microbiota composition. In this study, we investigated using a multi -omics strategy the changes in the gut microbiome induced by AML intense therapy and their association with gut barrier function and cachectic hallmarks. METHODS 10 AML patients, allocated to standard induction chemotherapy (SIC), were recruited. Samples and data were collected before any therapeutic intervention (T0), at the end of the SIC (T1) and at discharge (T4). Gut microbiota composition and function, markers of inflammation, metabolism, gut barrier function and cachexia, as well as faecal, blood and urine metabolomes were assessed. RESULTS AML patients demonstrated decreased appetite, weight loss and muscle wasting during hospitalization, with an incidence of cachexia of 50%. AML intensive treatment transiently impaired the gut barrier function and led to a long-lasting change of gut microbiota composition characterized by an important loss of diversity. Lactobacillaceae and Campylobacter concisus were increased at T1 while Enterococcus faecium and Staphylococcus were increased at T4. Metabolomics analyses revealed a reduction in urinary hippurate and faecal bacterial amino acid metabolites (bAAm) (2-methylbutyrate, isovalerate, phenylacetate). Integration using DIABLO revealed a deep interconnection between all the datasets. Importantly, we identified bacteria which disappearance was associated with impaired gut barrier function (Odoribacter splanchnicus) and body weight loss (Gemmiger formicilis), suggesting these bacteria as actionable targets. CONCLUSION AML intensive therapy transiently impairs the gut barrier function while inducing enduring alterations in the composition and metabolic activity of the gut microbiota that associate with body weight loss. TRIAL REGISTRATION NCT03881826, https://clinicaltrials.gov/ct2/show/NCT03881826.
Collapse
Affiliation(s)
- Sarah A Pötgens
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Sophie Lecop
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Violaine Havelange
- Department of Hematology, Cliniques Universitaires Saint-Luc, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Experimental Medicine Unit, De Duve Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Fuyong Li
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie Neveux
- Clinical Chemistry Department, Cochin Hospital, Paris Centre University Hospitals, Paris, France
| | - Johan Maertens
- Department of Hematology, University Hospital Gasthuisberg, Leuven, Belgium; Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jens Walter
- Department of Medicine, School of Microbiology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hélène Schoemans
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium; Department of Public Health and Primary Care, ACCENT VV, KU Leuven - University of Leuven, Leuven, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium; Welbio Department, WEL Research Institute, Wavre, Belgium.
| |
Collapse
|
3
|
Li Y, Zhou X, Lyu Z. Analysis of two-gene signatures and related drugs in small-cell lung cancer by bioinformatics. Open Med (Wars) 2023; 18:20230806. [PMID: 37808164 PMCID: PMC10560035 DOI: 10.1515/med-2023-0806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
Small-cell lung cancer (SCLC) has a poor prognosis and can be diagnosed with systemic metastases. Nevertheless, the molecular mechanisms underlying the development of SCLC are unclear, requiring further investigation. The current research aims to identify relevant biomarkers and available drugs to treat SCLC. The bioinformatics analysis comprised three Gene Expression Omnibus datasets (including GSE2149507, GSE6044, and GSE30219). Using the limma R package, we discovered differentially expressed genes (DEGs) in the current work. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were made by adopting the DAVID website. The DEG protein-protein interaction network was built based on the Search Tool for the Retrieval of Interacting Genes/Proteins website and visualized using the CytoHubba plugin in Cytoscape, aiming to screen the top ten hub genes. Quantitative real-time polymerase chain reaction was adopted for verifying the level of the top ten hub genes. Finally, the potential drugs were screened and identified using the QuartataWeb database. Totally 195 upregulated and 167 downregulated DEGs were determined. The ten hub genes were NCAPG, BUB1B, TOP2A, CCNA2, NUSAP1, UBE2C, AURKB, RRM2, CDK1, and KIF11. Ten FDA-approved drugs were screened. Finally, two genes and related drugs screened could be the prospective drug targets for SCLC treatment.
Collapse
Affiliation(s)
- Yi Li
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Xiwen Zhou
- Medical College, Shantou University, Shantou, China
| | - Zhi Lyu
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
- Department of Senior Cadres Ward, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Changes of Cell Adhesion Molecules and T Cell Subset Populations in Acute Myeloid Leukemia Patients Undergoing Intravenous Administration of Cytarabine Supplemented with Idarubicin. J CHEM-NY 2022. [DOI: 10.1155/2022/5507328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Objective. The present study aimed at investigating the efficacy and safety of intravenous administration of cytarabine supplemented with idarubicin in treating acute myeloid leukemia (AML) patients undergoing first attack and its effects on serum levels of cell adhesion molecules, cytokines in response to inflammation, and T cell subset populations in acute myeloid leukemia (AML) patients undergoing first attack. Methods. A total of 88 AML patients eligible for inclusion and exclusion criteria participated in the study and were randomly assigned into the control group (n = 44) in which the patients received intravenous administration of cytarabine and daunorubicin and the study group (n = 44) in which the patients received intravenous administration of cytarabine and idarubicin. Clinical response, incidence of adverse reactions, and quality of life 3 months after therapy were evaluated. Soluble intercellular adhesion molecule-1 (sICAM-1), soluble vascular cell adhesion molecule-1 (sVCAM-1), IL-10, and IL-35 were measured by ELISA methods. Phenotypic characteristics of T cell subsets including CD4+, CD8+, CD4+IL-10 Tregs, and CD4+CD25+CD127−Foxp3+ Tregs were analyzed by flow cytometry. Results. The clinical response rate of the study group was better than that of the control group (65.91% vs. 45.45%) (
). After treatment, the study group revealed significantly lower levels of sICAM-1, sVCAM-1, IL-10, and IL-35, a lower proportion of Tregs, a higher rate of CD4+/CD8+ T cells, along with increased scores of the Karnofsky Performance Scale (KPS) compared with the control group (
). The incidence rate of adverse reactions in the study group was lower than that in the control group (34.09% vs. 61.36%) (
). Conclusion. These findings demonstrate that intravenous administration of cytarabine supplemented with idarubicin can improve the immune function and quality of life of AML patients, and this combination drug therapy is effective and safe for AML.
Collapse
|
5
|
Zhang Q, Zhang CH, Wang ZD, Wang D. Efficacy and safety of induction chemotherapy with daunorubicin or idarubicin in the treatment of an adult with acute lymphoblastic leukemia. TUMORI JOURNAL 2021; 108:182-188. [PMID: 34296648 DOI: 10.1177/03008916211032724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Anthracyclines are dispensable components of chemotherapy of patients with acute lymphoblastic leukemia (ALL). OBJECTIVE To analyze the efficacy and safety of induction with idarubicin (IDA) or liposoma daunorubicin (L-DNR) in treatment of adults with high-risk ALL (HR-ALL) (presence of mixed lineage leukemia gene [MLL] rearrangements, t[1;19], or prednisone poor response). METHODS Among 58 enrolled patients, 29 cases were defined as the IDA group and the other 29 patients were put into the L-DNR group. Both overall survival (OS) and progression-free survival (PFS) were estimated and overall response rate (ORR) was compared between the groups. RESULTS The L-DNR group's OS and PFS were insignificantly higher than in the IDA group (P=0.261 and P=0.247). Although not significantly different, the ORR of adults with HR-ALL receiving L-DNR regimens was also higher than in the IDA group (P=0.085). Comprehensive cytogenetic analysis revealed that patients harboring MLL rearrangement, E2A-PBX1, and P53 mutation had poorer prognosis than others. All 58 patients experienced hematologic response in this study; however, the length of hematologic response in the IDA group was significantly longer than in the L-DNR group (P=0.005). The incidence of bleeding and infection was without significant difference between the groups (both P>0.05). CONCLUSION L-DNR proved to be an effective drug within a multiagent approach, which shows a favorable overall profile, as well as similar adverse events when compared with IDA in HR-ALL. Patients with E2A-PBX1 are much more sensitive to L-DNR than IDA. Despite some progress made, outcomes in MLL rearrangement or P53 mutation carriers remain unsatisfactory, and intensive treatment will be critical.
Collapse
Affiliation(s)
- Qi Zhang
- Publicity Section, The Second Affiliated Hospital of Mudanjiang University, Mudanjiang, China
| | - Chun-Hong Zhang
- Department of Urology Surgery, The Second Affiliated Hospital of Mudanjiang University, Mudanjiang, China
| | - Zhen-Dong Wang
- Department of ENT surgery, The Second Affiliated Hospital of Mudanjiang University, Mudanjiang, China
| | - Dong Wang
- Department of Hematology and Rheumatology, The Second Affiliated Hospital of Mudanjiang University, Mudanjiang, China
| |
Collapse
|
6
|
Ferraris D, Lapidus R, Truong P, Bollino D, Carter-Cooper B, Lee M, Chang E, LaRossa-Garcia M, Dash S, Gartenhaus R, Choi EY, Kipe O, Lam V, Mason K, Palmer R, Williams E, Ambulos N, Kamangar F, Zhang Y, Kapadia B, Jing Y, Emadi A. Pre-Clinical Activity of Amino-Alcohol Dimeric Naphthoquinones as Potential Therapeutics for Acute Myeloid Leukemia. Anticancer Agents Med Chem 2021; 22:239-253. [PMID: 34080968 DOI: 10.2174/1871520621666210602131558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 04/03/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The clinical outcomes of patients with acute myeloid leukemia (AML) remain unsatisfactory, therefore the development of more efficacious and better-tolerated therapy for AML is critical. We have previously reported the anti-leukemic activity of synthetic halohydroxyl dimeric naphthoquinones (BiQ) and aziridinyl BiQ. OBJECTIVE This study aimed to improve the potency and bioavailability of BiQ compounds and investigate the anti-leukemic activity of the lead compound in vitro and in a human AML xenograft mouse model. METHODS We designed, synthesized, and performed structure-activity relationship of several rationally designed BiQ analogues that possess amino alcohol functional groups on the naphthoquinone core rings. The compounds were screened for anti-leukemic activity and the mechanism as well as in vivo tolerability and efficacy of our lead compound was investigated. RESULTS We report that a dimeric naphthoquinone (designated BaltBiQ) demonstrated potent nanomolar anti-leukemic activity in AML cell lines. BaltBiQ treatment resulted in the generation of reactive oxygen species, induction of DNA damage, and inhibition of indoleamine dioxygenase 1. Although BaltBiQ was tolerated well in vivo, it did not significantly improve survival as a single agent, but in combination with the specific Bcl-2 inhibitor, Venetoclax, tumor growth was significantly inhibited compared to untreated mice. CONCLUSION We synthesized a novel amino alcohol dimeric naphthoquinone, investigated its main mechanisms of action, reported its in vitro anti-AML cytotoxic activity, and showed its in vivo promising activity combined with a clinically available Bcl-2 inhibitor in a patient-derived xenograft model of AML.
Collapse
Affiliation(s)
- Dana Ferraris
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Rena Lapidus
- University of Maryland School of Medicine, Morgan State University, Baltimore, MD, United States
| | - Phuc Truong
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Dominique Bollino
- University of Maryland School of Medicine, Morgan State University, Baltimore, MD, United States
| | - Brandon Carter-Cooper
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Michelle Lee
- University of Maryland School of Medicine, Morgan State University, Baltimore, MD, United States
| | - Elizabeth Chang
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Maria LaRossa-Garcia
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Smaraki Dash
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Ronald Gartenhaus
- Hunter Holmes McGuire Veterans Affairs Medical Center and Virginia Commonwealth University School of Medicine Department of Internal Medicine, Richmond, VA, United States
| | - Eun Yong Choi
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Olivia Kipe
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Vi Lam
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Kristopher Mason
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Riley Palmer
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Elijah Williams
- McDaniel College Department of Chemistry, 2 College Hill, Westminster, United States
| | - Nicholas Ambulos
- University of Maryland School of Medicine, Morgan State University, Baltimore, MD, United States
| | - Farin Kamangar
- Hunter Holmes McGuire Veterans Affairs Medical Center and Virginia Commonwealth University School of Medicine Department of Internal Medicine, Richmond, VA, United States
| | - Yuji Zhang
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Bandish Kapadia
- Hunter Holmes McGuire Veterans Affairs Medical Center and Virginia Commonwealth University School of Medicine Department of Internal Medicine, Richmond, VA, United States
| | - Yin Jing
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Ashkan Emadi
- University of Maryland School of Medicine, Morgan State University, Baltimore, MDun, United States
| |
Collapse
|
7
|
Kolesnikova MA, Sen'kova AV, Pospelova TI, Zenkova MA. Drug responsiveness of leukemic cells detected in vitro at diagnosis correlates with therapy response and survival in patients with acute myeloid leukemia. Cancer Rep (Hoboken) 2021; 4:e1362. [PMID: 33675187 PMCID: PMC8388166 DOI: 10.1002/cnr2.1362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/29/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is the most common acute leukemia in adults, and chemotherapy remains the most commonly used treatment approach for this group of hematological disorders. Drug resistance is one of the predictors of unfavorable prognosis for leukemia patients. AIM The purpose of this study was to perform a retrospective analysis of the survival rate in AML patients according to age, tumor status, and chemotherapy regimen received and to analyze the therapy response of AML patients depending on the treatment received, initial responsiveness of tumor cells to chemotherapeutic drugs measured in vitro at diagnosis and expression of immunological markers. METHODS The survival of AML patients (n = 127) was analyzed using the Kaplan-Meier method. Drug sensitivity of tumor cells of AML patients (n = 37) and the expression of immunological markers were evaluated by the WST test and flow cytometry, respectively. Correlation analysis was performed using Spearman's rank order correlation coefficient. RESULTS We found the treatment regimen to be the defining factor in the patient survival rate. In addition, the initial responsiveness of tumor cells to chemotherapeutic drugs measured in vitro at diagnosis correlated with the therapy response of AML: patients with high tumor cell sensitivity to particular cytotoxic drugs demonstrated a good response to treatment including these drugs, and patients with initial resistance of tumor cells to a particular chemotherapeutic agents and received it according to the clinical protocols demonstrated a poor response to antitumor therapy. Correlations of drug resistance in leukemic cells with the expression of immature and aberrant immunophenotype markers as established unfavorable prognostic factors confirm our assumption. CONCLUSION The evaluation of the responsiveness of tumor cells to chemotherapy in vitro at diagnosis can be a useful tool for predicting the response of leukemia patients to planned chemotherapy.
Collapse
Affiliation(s)
- Maria A Kolesnikova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia.,Novosibirsk Hematology Center, Novosibirsk, Russia
| | | | | | - Marina A Zenkova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| |
Collapse
|
8
|
Effect of Dose Ratio on Mitoxantrone and Daunorubicin in Acute Myeloid Leukemia: A Systematic Review and Meta-analysis of Randomized Controlled Trials. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:e10-e20. [PMID: 32863193 DOI: 10.1016/j.clml.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 11/20/2022]
Abstract
PURPOSE To investigate the effects of mitoxantrone and daunorubicin in induced chemotherapy on complete remission (CR), death during induction therapy, overall survival (OS), disease-free survival (DFS), and relapse in patients of all ages with acute myeloid leukemia (AML). METHODS We searched published reports at the Medline, Embase, and Cochrane Databases as well as other databases from inception through July 2019. There was no restriction on date of publication or language (PROSPERO registration CRD42018095843). RESULTS We enrolled 12 randomized controlled trials that included data of 4583 AML patients whose disease was untreated or relapsed/refractory, and compared the CR, death during induction therapy, DFS, and OS between mitoxantrone and daunorubicin. Mitoxantrone significantly increased the CR rate (relative risk = 1.07; 95% confidence interval [CI], 1.01, 1.14; P = .03) and DFS (hazard ratio = 0.87; 95% CI, 0.79, 0.96; P = .005) compared to daunorubicin. However, there was no significant difference in death during induction therapy (relative risk = 1.00; 95% CI, 0.81, 1.24; P = .99) and OS (hazard ratio = 0.94; 95% CI, 0.87, 1.01; P = .077) between the two drugs. CONCLUSION Although more studies are needed to compare mitoxantrone with higher-dose daunorubicin, the results showed that compared to daunorubicin, mitoxantrone can significantly improve CR and DFS in patients of all ages. These findings suggest that mitoxantrone may be a better choice than daunorubicin as an induction chemotherapy agent for AML patients, especially in developing countries.
Collapse
|
9
|
Wang H, Xiao X, Xiao Q, Lu Y, Wu Y. The efficacy and safety of daunorubicin versus idarubicin combined with cytarabine for induction therapy in acute myeloid leukemia: A meta-analysis of randomized clinical trials. Medicine (Baltimore) 2020; 99:e20094. [PMID: 32541448 PMCID: PMC7302600 DOI: 10.1097/md.0000000000020094] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE To ascertain the efficacy and safety of daunorubicin combined with cytarabine comparing with idarubicin combined with cytarabine as a standard induction therapy for acute Myeloid leukemia by a meta-analysis. METHODS The randomized controlled trials included were retrieved from PubMed, Embase, and Cochrane library. We evaluated and cross-checked the randomized clinical trials (RCTs) comparing daunorubicin combined with cytarabine (DA) and idarubicin combined with cytarabine (IA) by two reviewers independently according to Cochrane Handbook for Systematic Reviewers of Interventions. The data of meta-analysis was conducted using Review Manager 5.3 and Stata 12.0 software. RESULTS A total of 6 studies containing 3140 patients were included. The primary outcomes were complete remission (CR), CR in one course (CR1), CR in two courses (CR2), overall survival (OS), and relapse rate. The secondary outcomes included adverse events and cytogenetic risk in subgroup analyses. IA showed a statistically significant in CR (RR = 1.05; 95%CI = 1.00-1.09, P = .03) and CR1 (RR = 1.11; 95%CI = 1.04-1.18, P = .003), but not in CR2 (RR = 0.97; 95%CI = 0.77-1.24, P = .83), and relapse rate (RR = 1.08; 95%CI = 0.98-1.43, P = .08). In high dose daunorubicin group, OS was significantly improved with IA compared to DA (HR = 0.89, 95%CI = 0.8-1.0, P = .041, I = 0). At grade 3/4 adverse events, the difference between IA and DA was not statistically significant (infection, P = .28; cardiac toxicity, P = .15; bleeding, P = .29). In the subgroup analysis, the genotypes of the IA and DA groups were not statistically significant for comparison of CR between the two groups (P = .07). CONCLUSION This meta-analysis showed that IA had a better efficacy in the treatment of acute myeloid leukemia than DA, even with increased doses of DA. The OS of a standard dose of IA patients was longer than that of DA patients. Our research shows that anthracycline dose intensification of daunorubicin is of no clinically relevant benefit in AML patients comparing with a standard dose of IA. When it comes to adverse drug reactions, it is not a significant difference. Therefore, in clinical practice, IA should be the first choice for induction regimen in patients with acute myeloid leukemia.
Collapse
|