1
|
Ha H, Jeong Y, Lim JH, Suh YJ. Treatment Pattern, Financial Burden and Outcomes in Elderly Patients with Acute Myeloid Leukemia in Korea: A Nationwide Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042317. [PMID: 35206499 PMCID: PMC8872510 DOI: 10.3390/ijerph19042317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 12/10/2022]
Abstract
Although approximately 50% of patients with acute myeloid leukemia (AML) are diagnosed over the age of 60 years, there is currently no established consensus on the treatment of elderly AML patients. Herein, we aimed to explore the incidence, medical expenditure, treatment, and outcomes of elderly AML patients in Korea by analyzing a nationwide cohort. We employed the Korean National Health Insurance Service-Senior cohort, which represents 10% of a random selection from a total of 5.5 million subjects aged 60 years or older. AML patients were identified according to the main diagnostic criteria of acute leukemia. Treatment for AML was divided into high- (high-dose cytarabine ± idarubicin) and low- (low-dose cytarabine or hypomethylating agents) intensity chemo-therapy and classified according to the chemotherapeutics protocol. We analyzed the survival outcomes and medical expenditures. Among 558,147 elderly patients, 471 were diagnosed with AML, and 195 (41.4%) were treated with chemotherapy. The median age was 65 years, and the median overall survival (OS) was 4.93 months (95% confidence interval, 4.47–5.43). Median OS was longer in patients undergoing chemotherapy than those in the best supportive care group (6.28 vs. 3.45 months, p < 0.001), and the difference was prominent in patients aged < 70 years. Twenty-eight (5.9%) patients received high-intensity chemotherapy, while 146 (31.0%) received low-intensity chemotherapy. The difference in median OS according to dose intensity was 4.6 months, which was longer in the high-intensity chemotherapy group (9.8 vs. 5.2 months in low-intensity group); however, the difference was not statistically significant. Patients who received high-intensity chemotherapy recorded longer hospital stays and incurred greater expenses on initial hospitalization. Elderly AML patients in Korea exhibited clinical benefits from chemotherapy. Although patients should be carefully selected for intensive treatment, chemotherapy, including low-intensity treatment, can be considered in elderly patients. Moreover, prospective studies on new agents or new treatment strategies are needed.
Collapse
Affiliation(s)
- Hyerim Ha
- Department of Internal Medicine, Inha University Hospital, Incheon 22332, Korea;
| | - Yujin Jeong
- Department of Biostatistics, Korea University College of Medicine, Seoul 02841, Korea;
| | - Joo Han Lim
- Department of Internal Medicine, Inha University Hospital, Incheon 22332, Korea;
- Correspondence: (J.H.L.); (Y.J.S.); Tel.: +82-32-890-2581 (J.H.L.); +82-32-890-2833 (Y.J.S.)
| | - Young Ju Suh
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22332, Korea
- Correspondence: (J.H.L.); (Y.J.S.); Tel.: +82-32-890-2581 (J.H.L.); +82-32-890-2833 (Y.J.S.)
| |
Collapse
|
2
|
Colunga-Lozano LE, Kenji Nampo F, Agarwal A, Desai P, Litzow M, Sekeres MA, Guyatt GH, Brignardello-Petersen R. Less intensive antileukemic therapies (monotherapy and/or combination) for older adults with acute myeloid leukemia who are not candidates for intensive antileukemic therapy: A systematic review and meta-analysis. PLoS One 2022; 17:e0263240. [PMID: 35108310 PMCID: PMC8809589 DOI: 10.1371/journal.pone.0263240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 01/17/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Elderly patients with acute myeloid leukemia not eligible for intensive antileukemic therapy are treated with less intensive therapies, uncertainty remains regarding their relative merits. Objectives To compare the effectiveness and safety of less intensive antileukemic therapies for older adults with newly diagnosed AML not candidates for intensive therapies. Methods We included randomized controlled trials (RCTs) and non-randomized studies (NRS) comparing less intensive therapies in adults over 55 years with newly diagnosed AML. We searched MEDLINE and EMBASE from inception to August 2021. We assessed risk of bias of RCTs with a modified Cochrane Risk of Bias tool, and NRS with the Non-Randomized Studies of Interventions tool (ROBINS-I). We calculated pooled hazard ratios (HRs), risk ratios (RRs), mean differences (MD) and their 95% confidence intervals (CIs) using a random-effects pairwise meta-analyses and assessed the certainty of evidence using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. Results We included 27 studies (17 RCTs, 10 NRS; n = 5,698), which reported 9 comparisons. Patients were treated with azacitidine, decitabine, and low-dose cytarabine (LDAC), as monotherapies or in combination with other agents. Moderate certainty of evidence suggests no convincing difference in overall survival of patients who receive azacitidine monotherapy compared to LDAC monotherapy (HR 0.69; 95% CI, 0.31–1.53), fewer febrile neutropenia events occurred between azacitidine monotherapy to azacitidine combination (RR 0.45; 95% CI, 0.31–0.65), and, fewer neutropenia events occurred between LDAC monotherapy to decitabine monotherapy (RR 0.62; 95% CI 0.44–0.86). All other comparisons and outcomes had low or very low certainty of evidence. Conclusion There is no convincing superiority in OS when comparing less intensive therapies. Azacitidine monotherapy is likely to have fewer adverse events than azacitidine combination (febrile neutropenia), and LDAC monotherapy is likely to have fewer adverse events than decitabine monotherapy (neutropenia).
Collapse
Affiliation(s)
- Luis Enrique Colunga-Lozano
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| | - Fernando Kenji Nampo
- Department of Latin-American Institute of Life and Nature science, University of Latin-American Integration, Foz Do Iguaçu, Parana, Brazil
| | - Arnav Agarwal
- Department of Medicine, Toronto University, Toronto, Ontario, Canada
| | - Pinkal Desai
- Division of Hematology and Medical Oncology, Weill Cornell Medical Center, New York, New York, United States of America
| | - Mark Litzow
- Division of Hematology, Mayo clinic, Rochester, Minnesota, United states of America
| | - Mikkael A. Sekeres
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, United States of America
| | - Gordon H. Guyatt
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
3
|
Oliva EN, Ronnebaum SM, Zaidi O, Patel DA, Nehme SA, Chen C, Almeida AM. A systematic literature review of disease burden and clinical efficacy for patients with relapsed or refractory acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:325-360. [PMID: 34540343 PMCID: PMC8446831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a rapidly progressive hematological malignancy that is difficult to cure. The prognosis is poor and treatment options are limited in case of relapse. A comprehensive assessment of current disease burden and the clinical efficacy of non-intensive therapies in this population are lacking. We conducted two systematic literature reviews (SLRs). The first SLR (disease burden) included observational studies reporting the incidence and economic and humanistic burden of relapsed/refractory (RR) AML. The second SLR (clinical efficacy) included clinical trials (phase II or later) reporting remission rates (complete remission [CR] or CR with incomplete hematologic recovery [CRi]) and median overall survival (mOS) in patients with RR AML or patients with de novo AML who are ineligible for intensive chemotherapy. For both SLRs, MEDLINE®/Embase® were searched from January 1, 2008 to January 31, 2020. Clinical trial registries were also searched for the clinical efficacy SLR. After screening, two independent reviewers determined the eligibility for inclusion in the SLRs based on full-text articles. The disease burden SLR identified 130 observational studies. The median cumulative incidence of relapse was 29.4% after stem cell transplant and 46.8% after induction chemotherapy. Total per-patient-per-month costs were $28,148-$29,322; costs and health care resource use were typically higher for RR versus non-RR patients. Patients with RR AML had worse health-related quality of life (HRQoL) scores than patients with de novo AML across multiple instruments, and lower health utility values versus other AML health states (i.e. newly diagnosed, remission, consolidation, and maintenance therapy). The clinical efficacy SLR identified 50 trials (66 total trial arms). CR/CRi rates and mOS have remained relatively stable and low over the last 2 decades. Across all arms, the median rate of CR/CRi was 18.3% and mOS was 6.2 months. In conclusion, a substantial proportion of patients with AML will develop RR AML, which is associated with significant humanistic and economic burden. Existing treatments offer limited efficacy, highlighting the need for more effective non-intensive treatment options.
Collapse
|
4
|
Dennis M, Burnett A, Hills R, Thomas I, Ariti C, Severinsen MT, Hemmaway C, Greaves P, Clark RE, Copland M, Russell N. A randomised evaluation of low-dose cytosine arabinoside (ara-C) plus tosedostat versus low-dose ara-C in older patients with acute myeloid leukaemia: results of the LI-1 trial. Br J Haematol 2021; 194:298-308. [PMID: 33961292 DOI: 10.1111/bjh.17501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 11/26/2022]
Abstract
Older patients with acute myeloid leukaemia (AML) account for nearly half of those with the disease. Because they are perceived to be unfit for, unwilling to receive, or unlikely to benefit from conventional chemotherapy they represent an important unmet need. Tosedostat is a selective oral aminopeptidase inhibitor, which in phase I/II trials showed acceptable toxicity and encouraging efficacy. We report the only randomised study of low-dose cytosine arabinoside (LDAC) combined with tosedostat (LDAC-T) versus LDAC in untreated older patients not suitable for intensive treatment. A total of 243 patients were randomised 1:1 as part of the 'Pick-a-Winner' LI-1 trial. There was a statistically non-significant increase in the complete remission (CR) rate with the addition of tosedostat, LDAC-T 19% versus LDAC 12% [odds ratio (OR) 0·61, 95% confidence interval (CI) 0·30-1·23; P = 0·17]. For overall response (CR+CR with incomplete recovery of counts), there was little evidence of a benefit to the addition of tosedostat (25% vs. 18%; OR 0·68, 95% CI 0·37-1·27; P = 0·22). However, overall survival (OS) showed no difference (2-year OS 16% vs. 12%, hazard ratio 0·97, 95% CI 0·73-1·28; P = 0·8). Exploratory analyses failed to identify any subgroup benefitting from tosedostat. Despite promising pre-clinical, early non-randomised clinical data with acceptable toxicity and an improvement in response, we did not find evidence that the addition of tosedostat to LDAC produced a survival benefit in this group of patients with AML. International Standard Randomised Controlled Trial Number: ISRCTN40571019.
Collapse
Affiliation(s)
- Mike Dennis
- Department of Haematology, Christie Hospital NHS Trust, Manchester, UK
| | - Alan Burnett
- Paul O'Gorman Leukaemia Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Robert Hills
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ian Thomas
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Cono Ariti
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Marianne T Severinsen
- Department of Haematology, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Claire Hemmaway
- Department of Haematology, Auckland City Hospital, Auckland, New Zealand
| | - Paul Greaves
- Department of Haematology, Queen's Hospital, Romford, UK
| | - Richard E Clark
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Mhairi Copland
- Paul O'Gorman Leukaemia Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Nigel Russell
- Department of Haematology, Nottingham University Hospitals, Nottingham, UK
| |
Collapse
|
5
|
Kumar S, Nagpal R, Kumar A, Ashraf MU, Bae YS. Immunotherapeutic Potential of m6A-Modifiers and MicroRNAs in Controlling Acute Myeloid Leukaemia. Biomedicines 2021; 9:690. [PMID: 34207299 PMCID: PMC8234128 DOI: 10.3390/biomedicines9060690] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/30/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Epigenetic alterations have contributed greatly to human carcinogenesis. Conventional epigenetic studies have been predominantly focused on DNA methylation, histone modifications, and chromatin remodelling. Epitranscriptomics is an emerging field that encompasses the study of RNA modifications that do not affect the RNA sequence but affect functionality via a series of RNA binding proteins called writer, reader and eraser. Several kinds of epi-RNA modifications are known, such as 6-methyladenosine (m6A), 5-methylcytidine (m5C), and 1-methyladenosine. M6A modification is the most studied and has large therapeutic implications. In this review, we have summarised the therapeutic potential of m6A-modifiers in controlling haematological disorders, especially acute myeloid leukaemia (AML). AML is a type of blood cancer affecting specific subsets of blood-forming hematopoietic stem/progenitor cells (HSPCs), which proliferate rapidly and acquire self-renewal capacities with impaired terminal cell-differentiation and apoptosis leading to abnormal accumulation of white blood cells, and thus, an alternative therapeutic approach is required urgently. Here, we have described how RNA m6A-modification machineries EEE (Editor/writer: Mettl3, Mettl14; Eraser/remover: FTO, ALKBH5, and Effector/reader: YTHDF-1/2) could be reformed into potential druggable candidates or as RNA-modifying drugs (RMD) to treat leukaemia. Moreover, we have shed light on the role of microRNAs and suppressors of cytokine signalling (SOCS/CISH) in increasing anti-tumour immunity towards leukaemia. We anticipate, our investigation will provide fundamental knowledge in nurturing the potential of RNA modifiers in discovering novel therapeutics or immunotherapeutic procedures.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea;
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA;
| | - Amit Kumar
- Medical Writer, Quebec City, QC G1X 3E1, Canada;
| | - Muhammad Umer Ashraf
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea;
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea;
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| |
Collapse
|
6
|
Tsai HJ, Hsiao HH, Hsu YT, Liu YC, Kao HW, Liu TC, Cho SF, Feng X, Johnston A, Bomalaski JS, Kuo MC, Chen TY. Phase I study of ADI-PEG20 plus low-dose cytarabine for the treatment of acute myeloid leukemia. Cancer Med 2021; 10:2946-2955. [PMID: 33787078 PMCID: PMC8085967 DOI: 10.1002/cam4.3871] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Most acute myeloid leukemia (AML) cells are argininosuccinate synthetase‐deficient. Pegylated arginine deiminase (ADI‐PEG20) monotherapy depletes circulating arginine, thereby selectively inducing tumor cell death. ADI‐PEG20 was shown to induce complete responses in ~10% of relapsed/refractory or poor‐risk AML patients. We conducted a phase I, dose‐escalation study combining ADI‐PEG20 and low‐dose cytarabine (LDC) in AML patients. Patients received 20 mg LDC subcutaneously twice daily for 10 days every 28 days and ADI‐PEG20 at 18 or 36 mg/m2 (dose levels 1 and 2) intramuscularly weekly. An expansion cohort for the maximal tolerated dose of ADI‐PEG20 was planned to further estimate the toxicity and preliminary response of this regimen. The primary endpoints were safety and tolerability. The secondary endpoints were time on treatment, overall survival (OS), overall response rate (ORR), and biomarkers (pharmacodynamics and immunogenicity detection). Twenty‐three patients were included in the study, and seventeen patients were in the expansion cohort (dose level 2). No patients developed dose‐limiting toxicities. The most common grade III/IV toxicities were thrombocytopenia (61%), anemia (52%), and neutropenia (30%). One had an allergic reaction to ADI‐PEG20. The ORR in 18 evaluable patients was 44.4%, with a median OS of 8.0 (4.5‐not reached) months. In seven treatment‐naïve patients, the ORR was 71.4% and the complete remission rate was 57.1%. The ADI‐PEG20 and LDC combination was well‐tolerated and resulted in an encouraging ORR. Further combination studies are warranted. (This trial was registered in ClinicalTrials.gov as a Ph1 Study of ADI‐PEG20 Plus Low‐Dose Cytarabine in Older Patients With AML, NCT02875093).
Collapse
Affiliation(s)
- Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.,Department of Oncology, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.,Division of Hematology/Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Hua Hsiao
- Division of Hematology/Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ya-Ting Hsu
- Division of Hematology, Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Chang Liu
- Division of Hematology/Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiao-Wen Kao
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan
| | - Ta-Chih Liu
- Division of Hematology/Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Hematology-Oncology and Cancer Center, Chang Bing Show Chwan Hospital, Changhua, Taiwan
| | - Shih-Feng Cho
- Division of Hematology/Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Xiaoxing Feng
- Polaris Pharmaceuticals, Inc, Polaris Group, San Diego, CA, USA
| | - Amanda Johnston
- Polaris Pharmaceuticals, Inc, Polaris Group, San Diego, CA, USA
| | | | - Ming-Chung Kuo
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Linkou, Taiwan
| | - Tsai-Yun Chen
- Division of Hematology, Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
7
|
BST-236, a novel cytarabine prodrug for patients with acute leukemia unfit for standard induction: a phase 1/2a study. Blood Adv 2020; 3:3740-3749. [PMID: 31770437 DOI: 10.1182/bloodadvances.2019000468] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
High-dose cytarabine is the backbone of acute myeloid leukemia (AML) treatment. Nevertheless, its use in older patients is considerably limited due to increased toxicity. BST-236 (INN aspacytarabine) is a novel cytarabine prodrug designed to deliver high-dose cytarabine to target cells with reduced systemic exposure to free cytarabine. This phase 1/2a dose-escalation study was designed to evaluate BST-236 safety, pharmacokinetics, and efficacy in older or unfit-for-intensive-therapy patients with acute leukemia. Twenty-six patients, unfit for standard therapy, who were either relapsed/refractory or newly diagnosed, received BST-236 in 6 dose-escalating cohorts (range 0.3 to 6 g/m2 per day). BST-236 was administered intravenously once daily over 60 minutes for 6 consecutive days. The median age was 76.5 (26 to 90), with 84.6% of patients ≥70 years. BST-236 was safe and well tolerated. The maximal tolerated dose was 6 g/m2 per day. Overall response rate was 29.6%. A subgroup analysis of newly diagnosed patients with AML, de novo or secondary to myelodysplastic syndrome, unfit for standard induction (median age 78), demonstrated overall response of 45.5%. The median overall survival was 6.5 months and was not reached in patients achieving complete remission. The findings of this phase 1/2 study suggest that BST-236 safely delivers high and efficacious cytarabine doses to older patients who are unfit for standard induction and lays the foundation for further studies of BST-236 in AML. This trial was registered at www.clinicaltrials.gov as #NCT02544438.
Collapse
|
8
|
Sekeres MA, Guyatt G, Abel G, Alibhai S, Altman JK, Buckstein R, Choe H, Desai P, Erba H, Hourigan CS, LeBlanc TW, Litzow M, MacEachern J, Michaelis LC, Mukherjee S, O'Dwyer K, Rosko A, Stone R, Agarwal A, Colunga-Lozano LE, Chang Y, Hao Q, Brignardello-Petersen R. American Society of Hematology 2020 guidelines for treating newly diagnosed acute myeloid leukemia in older adults. Blood Adv 2020; 4:3528-3549. [PMID: 32761235 PMCID: PMC7422124 DOI: 10.1182/bloodadvances.2020001920] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/08/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Older adults with acute myeloid leukemia (AML) represent a vulnerable population in whom disease-based and clinical risk factors, patient goals, prognosis, and practitioner- and patient-perceived treatment risks and benefits influence treatment recommendations. OBJECTIVE These evidence-based guidelines of the American Society of Hematology (ASH) are intended to support patients, clinicians, and other health care professionals in their decisions about management of AML in older adults. METHODS ASH formed a multidisciplinary guideline panel that included specialists in myeloid leukemia, geriatric oncology, patient-reported outcomes and decision-making, frailty, epidemiology, and methodology, as well as patients. The McMaster Grading of Recommendations Assessment, Development and Evaluation (GRADE) Centre supported the guideline-development process, including performing systematic evidence reviews (up to 24 May 2019). The panel prioritized clinical questions and outcomes according to their importance to patients, as judged by the panel. The panel used the GRADE approach, including GRADE's Evidence-to-Decision frameworks, to assess evidence and make recommendations, which were subject to public comment. RESULTS The panel agreed on 6 critical questions in managing older adults with AML, mirroring real-time practitioner-patient conversations: the decision to pursue antileukemic treatment vs best supportive management, the intensity of therapy, the role and duration of postremission therapy, combination vs monotherapy for induction and beyond, duration of less-intensive therapy, and the role of transfusion support for patients no longer receiving antileukemic therapy. CONCLUSIONS Treatment is recommended over best supportive management. More-intensive therapy is recommended over less-intensive therapy when deemed tolerable. However, these recommendations are guided by the principle that throughout a patient's disease course, optimal care involves ongoing discussions between clinicians and patients, continuously addressing goals of care and the relative risk-benefit balance of treatment.
Collapse
Affiliation(s)
- Mikkael A Sekeres
- Leukemia Program, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Gordon Guyatt
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Gregory Abel
- Leukemia Division, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Shabbir Alibhai
- Institute of Medical Sciences, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jessica K Altman
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Rena Buckstein
- Odette Cancer Centre, Division of Medical Oncology and Hematology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hannah Choe
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Pinkal Desai
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY
| | - Harry Erba
- Department of Medicine, School of Medicine, Duke University, Durham, NC
| | | | - Thomas W LeBlanc
- Department of Medicine, School of Medicine, Duke University, Durham, NC
| | - Mark Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Laura C Michaelis
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Sudipto Mukherjee
- Leukemia Program, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Kristen O'Dwyer
- Division of Hematology/Oncology, Department of Medicine, University of Rochester, Rochester, NY
| | - Ashley Rosko
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Richard Stone
- Leukemia Division, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Arnav Agarwal
- Department of Internal Medicine, University of Toronto, Toronto, ON, Canada
| | - L E Colunga-Lozano
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- Health Science Center, Department of Clinical Medicine, Universidad de Guadalajara, Guadalajara, Mexico; and
| | - Yaping Chang
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - QiuKui Hao
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- The Center of Gerontology and Geriatrics/National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
9
|
Updates on DNA methylation modifiers in acute myeloid leukemia. Ann Hematol 2020; 99:693-701. [DOI: 10.1007/s00277-020-03938-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Accepted: 01/24/2020] [Indexed: 12/14/2022]
|
10
|
Recent advances in the synthetic and medicinal perspective of quinolones: A review. Bioorg Chem 2019; 92:103291. [PMID: 31561107 DOI: 10.1016/j.bioorg.2019.103291] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022]
Abstract
In the modern scenario, the quinolone scaffold has emerged as a very potent motif considering its clinical significance. Quinolones possess wide range of pharmacological activities such as anticancer, antibacterial, antifungal, antiprotozoal, antiviral, anti-inflammatory, carbonic anhydrase inhibitory and diuretic activity etc. The versatile synthetic approaches have been successfully applied and several of the resulted synthesized compounds exhibit fascinating biological activities in numerous fields. This has prompted to discover quinolone-based analogues among the researchers due to its great diversity in biological activities. In the past few years, various new, efficient and convenient synthetic approaches (including green chemistry and microwave-assisted synthesis) have been designed and developed to synthesize diverse quinolone-based scaffolds which represent a growing area of interest in academic and industry as well as to explore their biological activities. In this review, an attempt has been made by the authors to summarize (1) One of the most comprehensive listings of quinolone-based drugs or agents in the market or under various stages of clinical development; (2) Recent advances in the synthetic strategies for quinolone derivatives as well as their biological implications including insight of mechanistic studies. (3) Further, the biological data is correlated with structure-activity relationship studies to provide an insight into the rational design of more active agents.
Collapse
|
11
|
Stone A, Zukerman T, Flaishon L, Yakar RB, Rowe JM. Efficacy outcomes in the treatment of older or medically unfit patients with acute myeloid leukaemia: A systematic review and meta-analysis. Leuk Res 2019; 82:36-42. [DOI: 10.1016/j.leukres.2019.05.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 01/03/2023]
|
12
|
Economides MP, McCue D, Borthakur G, Pemmaraju N. Topoisomerase II inhibitors in AML: past, present, and future. Expert Opin Pharmacother 2019; 20:1637-1644. [PMID: 31136213 DOI: 10.1080/14656566.2019.1621292] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Topoisomerase II inhibitors have long been used in the frontline and as salvage therapy for AML, with daunorubicin and idarubicin being prototypical agents in this therapeutic class, classically in combination with nucleoside analogs, e.g. cytarabine. Most recently, several other compounds from this drug class have or are being investigated. Areas covered: The current paper reviews older and newer topoisomerase II inhibitors in clinical development for the treatment of AML. The authors discuss the clinical use of these agents, current trials involving them as well as their safety profile. Important side effects of these medications including therapy-related AML (t-AML) are also covered. Expert opinion: Topoisomerase II inhibitors have helped improve outcomes in AML. Recently, the FDA approved several agents including CPX-351 for the treatment of secondary and t-AML. CPX-351 may have applicability in other high-risk myeloid diseases. Future directions include a combination of these agents with other targeted therapies. Finally, the authors believe that small molecule inhibitors, such as venetoclax and possibly immunotherapy options could also be incorporated to our treatment paradigm in selected patients.
Collapse
Affiliation(s)
- Minas P Economides
- Department of Internal Medicine, The University of Texas School of Health Sciences at Houston , Houston , TX , USA
| | - Deborah McCue
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
13
|
Venetoclax Synergistically Enhances the Anti-leukemic Activity of Vosaroxin Against Acute Myeloid Leukemia Cells Ex Vivo. Target Oncol 2019; 14:351-364. [DOI: 10.1007/s11523-019-00638-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Burnett AK. Treatment of Older Patients With Newly Diagnosed AML Unfit for Traditional Therapy. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 18:553-557. [PMID: 30007570 DOI: 10.1016/j.clml.2018.06.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 06/26/2018] [Indexed: 11/28/2022]
Abstract
Older patients with acute myeloid leukemia represent at least one half of those with the disease for whom randomized clinical trials of new treatments are in development. These patients represent an appropriate population in which to evaluate new treatments against the current standards of care, which could be azacitidine, decitabine, or low-dose cytarabine. However, despite the identification of treatments that can deliver a worthwhile increase in remission, none has yet delivered a survival superiority when assessed in a randomized setting, although some recent efforts provide encouragement.
Collapse
Affiliation(s)
- Alan K Burnett
- (Retired), Cardiff University Ty Mawr, Blackwaterfoot, United Kingdom.
| |
Collapse
|
15
|
Abstract
Acute myeloid leukemia (AML) therapies are rapidly evolving with novel targeted therapies showing high-level responses in a notoriously difficult to treat group of patients - the elderly and unfit. This review will examine the outcomes of older AML patients (>60 years old) with conventional induction strategies, and published literature on risks of pursuit of induction. Low-intensity combination therapy response rates appear to be approaching that of induction regimens, and with lower toxicity, low-intensity therapy likely represents the future standard approach in this age group. Lastly, allogeneic transplant appears to have a role in increasing durable remissions regardless of age and should be considered in patients with limited comorbidities.
Collapse
Affiliation(s)
- Jonathan A Webster
- a Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| | - Keith W Pratz
- a Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Although the treatment paradigm for acute myeloid leukemia (AML) had been largely unchanged for many years, in-depth molecular characterization has revolutionized our understanding of mutations that drive the disease, subsequently serving to guide current clinical investigation. Furthermore, recent advances in the field have highlighted the importance of optimizing known efficacious agents by improving drug delivery or bypassing resistance mechanisms. The current status of novel agents which are shaping the clinical management of AML patients are summarized in this review. RECENT FINDINGS Practice changing findings over the past year include improved overall survival (OS) in a molecularly defined AML subgroup as well as in elderly patients with secondary AML (sAML). Specifically, synergistic combination of daunorubicin and cytarabine (i.e., CPX-351) was found to improve OS in sAML patients. Furthermore, although multiple mutation specific inhibitors have been developed, optimal combination with additional agents appears critical, as monotherapies have not resulted in durable remissions or improved outcomes. Improved OS via the addition of midostaurin to intensive chemotherapy in FLT3 mutant AML supports this concept. SUMMARY For the first time in AML, personalized therapy has become possible through improved understanding of the molecular architecture and survival pathways of an individual's disease. The landscape of AML treatment is encouraging, with multiple novel agents likely to gain approval over the next 5 years.
Collapse
|
17
|
Sato A, Shimura M, Gosho M. Practical characteristics of adaptive design in phase 2 and 3 clinical trials. J Clin Pharm Ther 2017; 43:170-180. [PMID: 28850685 DOI: 10.1111/jcpt.12617] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 08/07/2017] [Indexed: 01/14/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Adaptive design methods are expected to be ethical, reflect real medical practice, increase the likelihood of research and development success and reduce the allocation of patients into ineffective treatment groups by the early termination of clinical trials. However, the comprehensive details regarding which types of clinical trials will include adaptive designs remain unclear. We examined the practical characteristics of adaptive design used in clinical trials. METHODS We conducted a literature search of adaptive design clinical trials published from 2012 to 2015 using PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials, with common search terms related to adaptive design. We systematically assessed the types and characteristics of adaptive designs and disease areas employed in the adaptive design trials. RESULTS AND DISCUSSION Our survey identified 245 adaptive design clinical trials. The number of trials by the publication year increased from 2012 to 2013 and did not greatly change afterwards. The most frequently used adaptive design was group sequential design (n = 222, 90.6%), especially for neoplasm or cardiovascular disease trials. Among the other types of adaptive design, adaptive dose/treatment group selection (n = 21, 8.6%) and adaptive sample-size adjustment (n = 19, 7.8%) were frequently used. The adaptive randomization (n = 8, 3.3%) and adaptive seamless design (n = 6, 2.4%) were less frequent. Adaptive dose/treatment group selection and adaptive sample-size adjustment were frequently used (up to 23%) in "certain infectious and parasitic diseases," "diseases of nervous system," and "mental and behavioural disorders" in comparison with "neoplasms" (<6.6%). For "mental and behavioural disorders," adaptive randomization was used in two trials of eight trials in total (25%). Group sequential design and adaptive sample-size adjustment were used frequently in phase 3 trials or in trials where study phase was not specified, whereas the other types of adaptive designs were used more in phase 2 trials. Approximately 82% (202 of 245 trials) resulted in early termination at the interim analysis. Among the 202 trials, 132 (54% of 245 trials) had fewer randomized patients than initially planned. This result supports the motive to use adaptive design to make study durations shorter and include a smaller number of subjects. WHAT IS NEW AND CONCLUSION We found that adaptive designs have been applied to clinical trials in various therapeutic areas and interventions. The applications were frequently reported in neoplasm or cardiovascular clinical trials. The adaptive dose/treatment group selection and sample-size adjustment are increasingly common, and these adaptations generally follow the Food and Drug Administration's (FDA's) recommendations.
Collapse
Affiliation(s)
- A Sato
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Novartis Pharma K.K., Tokyo, Japan
| | - M Shimura
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Data Science Department, Taiho Pharmaceutical Co. Ltd., Tokyo, Japan
| | - M Gosho
- Department of Clinical Trial and Clinical Epidemiology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
18
|
Kukowska M. Amino acid or peptide conjugates of acridine/acridone and quinoline/quinolone-containing drugs. A critical examination of their clinical effectiveness within a twenty-year timeframe in antitumor chemotherapy and treatment of infectious diseases. Eur J Pharm Sci 2017; 109:587-615. [PMID: 28842352 DOI: 10.1016/j.ejps.2017.08.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 08/16/2017] [Accepted: 08/19/2017] [Indexed: 01/10/2023]
Abstract
Acridines/acridones, quinolines/quinolones (chromophores) and their derivatives constitute extremely important family of compounds in current medicine. Great significance of the compounds is connected with antimicrobial and antitumor activities. Combining these features together in one drug seems to be long-term benefit, especially in oncology therapy. The attractiveness of the chromophore drugs is still enhanced by elimination their toxicity and improvement not only selectivity, specificity but also bioavailability. The best results are reached by conjugation to natural peptides. This paper highlights significant advance in the study of amino acid or peptide chromophore conjugates that provide highly encouraging data for novel drug development. The structures and clinical significance of amino acid or peptide chromophore conjugates are widely discussed.
Collapse
Affiliation(s)
- Monika Kukowska
- Chair & Department of Chemical Technology of Drugs, Faculty of Pharmacy with Subfaculty of Laboratory Medicine, Medical University of Gdansk, Al. Gen. J. Hallera 107, 80-416 Gdansk, Poland.
| |
Collapse
|
19
|
Abstract
OPINION STATEMENT Approximately 40-45% of younger and 10-20% of older adults with acute myeloid leukemia (AML) will be cured with current standard chemotherapy. The outlook is particularly gloomy for patients with relapsed and/or refractory disease (cure rates no higher than 10%). Allogeneic hematopoietic stem cell transplantation (HSCT), the only realistic hope of cure for these patients, is an option for only a minority. In recent years, much has been learned about the genomic and epigenomic landscapes of AML, and the clonal architecture of both de novo and secondary AML has begun to be unraveled. These advances have paved the way for rational drug development as new "drugable" targets have emerged. Although no new drug has been approved for AML in over four decades, with the exception of gemtuzumab ozogamycin, which was subsequently withdrawn, there is progress on the horizon with the possible regulatory approval soon of agents such as CPX-351 and midostaurin, the Food and Drug Administration "breakthrough" designation granted to venetoclax, and promising agents such as the IDH inhibitors AG-221 and AG-120, the smoothened inhibitor glasdegib and the histone deacetylase inhibitor pracinostat. In our practice, we treat most patients with relapsed/refractory AML on clinical trials, taking into consideration their prior treatment history and response to the same. We utilize targeted sequencing of genes frequently mutated in AML to identify "actionable" mutations, e.g., in FLT3 or IDH1/2, and incorporate small-molecule inhibitors of these oncogenic kinases into our therapeutic regimens whenever possible. In the absence of actionable mutations, we rationally combine conventional agents with other novel therapies such as monoclonal antibodies and other targeted drugs. For fit patients up to the age of 65, we often use high-dose cytarabine-containing backbone regimens. For older or unfit patients, we prefer hypomethylating agent-based therapy. Finally, all patients with relapsed/refractory AML are evaluated for allogeneic HSCT.
Collapse
|
20
|
Abstract
Vosaroxin, a quinolone-derivative chemotherapeutic agent, was considered a promising drug for the treatment of acute myeloid leukemia (AML). Early-stage clinical trials with this agent led to a large randomized double-blind placebo-controlled study of vosaroxin in combination with intermediate-dose cytarabine for the treatment of relapsed or refractory AML. The study demonstrated better complete remission rates with vosaroxin, but there was no statistically significant overall survival benefit in the whole cohort. A subset analysis censoring patients who had undergone allogeneic stem cell transplantation, however, revealed a modest but statistically significant improvement in overall survival particularly among older patients. This article reviews the data available on vosaroxin including clinical trials in AML and offers an analysis of findings of these studies as well as the current status of vosaroxin.
Collapse
Affiliation(s)
- Hamid Sayar
- Indiana University Simon Cancer Center, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
21
|
Jamieson GC, Fox JA, Poi M, Strickland SA. Molecular and Pharmacologic Properties of the Anticancer Quinolone Derivative Vosaroxin: A New Therapeutic Agent for Acute Myeloid Leukemia. Drugs 2017; 76:1245-1255. [PMID: 27484675 PMCID: PMC4989016 DOI: 10.1007/s40265-016-0614-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Vosaroxin is a first-in-class anticancer quinolone derivative that targets topoisomerase II and induces site-selective double-strand breaks in DNA, leading to tumor cell apoptosis. Vosaroxin has chemical and pharmacologic characteristics distinct from other topoisomerase II inhibitors due to its quinolone scaffold. The efficacy and safety of vosaroxin in combination with cytarabine were evaluated in patients with relapsed/refractory acute myeloid leukemia (AML) in a phase III, randomized, multicenter, double-blind, placebo-controlled study (VALOR). In this study, the addition of vosaroxin produced a 1.4-month improvement in median overall survival (OS; 7.5 months with vosaroxin/cytarabine vs. 6.1 months with placebo/cytarabine; hazard ratio [HR] 0.87, 95 % confidence interval [CI] 0.73−1.02; unstratified log-rank p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}= 0.061; stratified log-rank p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}=0.024), with the greatest OS benefit observed in patients ≥60 years of age (7.1 vs. 5.0 months; HR 0.75, 95 % CI 0.62−0.92; p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}=0.003) and patients with early relapse (6.7 vs. 5.2 months; HR 0.77, 95 % CI 0.59−1.00; p\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$=$$\end{document}= 0.039), two AML patient groups that typically have poor prognosis. Here we review the chemical and pharmacologic properties of vosaroxin, how these properties are distinct from those of currently available topoisomerase II inhibitors, how they may contribute to the efficacy and safety profile observed in the VALOR trial, and the status of clinical development of vosaroxin for treatment of AML.
Collapse
Affiliation(s)
| | - Judith A Fox
- Sunesis Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Ming Poi
- College of Pharmacy, Ohio State University, Columbus, OH, USA
| | - Stephen A Strickland
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN, 37232, USA.
| |
Collapse
|
22
|
Loke J, Assi SA, Imperato MR, Ptasinska A, Cauchy P, Grabovska Y, Soria NM, Raghavan M, Delwel HR, Cockerill PN, Heidenreich O, Bonifer C. RUNX1-ETO and RUNX1-EVI1 Differentially Reprogram the Chromatin Landscape in t(8;21) and t(3;21) AML. Cell Rep 2017; 19:1654-1668. [PMID: 28538183 PMCID: PMC5457485 DOI: 10.1016/j.celrep.2017.05.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/13/2017] [Accepted: 04/28/2017] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease caused by mutations in transcriptional regulator genes, but how different mutant regulators shape the chromatin landscape is unclear. Here, we compared the transcriptional networks of two types of AML with chromosomal translocations of the RUNX1 locus that fuse the RUNX1 DNA-binding domain to different regulators, the t(8;21) expressing RUNX1-ETO and the t(3;21) expressing RUNX1-EVI1. Despite containing the same DNA-binding domain, the two fusion proteins display distinct binding patterns, show differences in gene expression and chromatin landscape, and are dependent on different transcription factors. RUNX1-EVI1 directs a stem cell-like transcriptional network reliant on GATA2, whereas that of RUNX1-ETO-expressing cells is more mature and depends on RUNX1. However, both types of AML are dependent on the continuous expression of the fusion proteins. Our data provide a molecular explanation for the differences in clinical prognosis for these types of AML.
Collapse
Affiliation(s)
- Justin Loke
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - Salam A Assi
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - Maria Rosaria Imperato
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - Anetta Ptasinska
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - Pierre Cauchy
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - Yura Grabovska
- Northern Institute for Cancer Research, University of Newcastle, Newcastle upon Tyne NE2 4HH, UK
| | - Natalia Martinez Soria
- Northern Institute for Cancer Research, University of Newcastle, Newcastle upon Tyne NE2 4HH, UK
| | - Manoj Raghavan
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - H Ruud Delwel
- Department of Hematology, Erasmus University Medical Center, Dr. Molewaterplein 50, 3015 GE Rotterdam, the Netherlands
| | - Peter N Cockerill
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK
| | - Olaf Heidenreich
- Northern Institute for Cancer Research, University of Newcastle, Newcastle upon Tyne NE2 4HH, UK
| | - Constanze Bonifer
- Institute for Cancer and Genomic Sciences, College of Medicine and Dentistry, University of Birmingham, B15 2TT Birmingham, UK.
| |
Collapse
|
23
|
Paubelle E, Zylbersztejn F, Thomas X. The preclinical discovery of vosaroxin for the treatment of acute myeloid leukemia. Expert Opin Drug Discov 2017; 12:747-753. [PMID: 28504025 DOI: 10.1080/17460441.2017.1331215] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) represents a disease with a very poor outcome and remains an area of significant unmet need necessitating novel therapeutic strategies. Among novel therapeutic agents, vosaroxin is a first-in-class anticancer quinolone derivative that targets topoisomerase II and induces site-selective double-strand breaks in DNA, leading to tumor cell apoptosis. Areas covered: Herein, the authors provide a comprehensive review of the preclinical development of vosaroxin. This includes coverage of vosaroxin's mechanism of action in addition to its pharmacology and of the main studies reported over the past few years with vosaroxin when used to treat adult AML. Expert opinion: Given that vosaroxin is associated with fewer potential side effects, it may be of benefit to elderly patients with relapsed/refractory AML and to those with additional comorbidities who have previously received an anthracycline and cytarabine combination. Furthermore, vosaroxin also was seen to be active in multidrug-resistant preclinical models. However, further studies have to be performed to better evaluate its place in the armamentarium against AML.
Collapse
Affiliation(s)
- Etienne Paubelle
- a Hospices Civils de Lyon, Hematology Department , Lyon-Sud Hospital , Pierre-Bénite , France
| | | | - Xavier Thomas
- a Hospices Civils de Lyon, Hematology Department , Lyon-Sud Hospital , Pierre-Bénite , France
| |
Collapse
|
24
|
Reichenbach F, Wiedenmann C, Schalk E, Becker D, Funk K, Scholz-Kreisel P, Todt F, Wolleschak D, Döhner K, Marquardt JU, Heidel F, Edlich F. Mitochondrial BAX Determines the Predisposition to Apoptosis in Human AML. Clin Cancer Res 2017; 23:4805-4816. [DOI: 10.1158/1078-0432.ccr-16-1941] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/05/2016] [Accepted: 04/11/2017] [Indexed: 11/16/2022]
|
25
|
Abstract
Acute myeloid leukemia (AML) is characterized by clinical and biological heterogeneity. Despite the advances in our understanding of its pathobiology, the chemotherapy-directed management has remained largely unchanged in the past 40 years. However, various novel agents have demonstrated clinical activity, either as single agents (e.g., isocitrate dehydrogenase (IDH) inhibitors, vadastuximab) or in combination with standard induction/consolidation at diagnosis and with salvage regimens at relapse. The classes of agents described in this review include novel cytotoxic chemotherapies (CPX-351 and vosaroxin), epigenetic modifiers (guadecitabine, IDH inhibitors, histone deacetylase (HDAC) inhibitors, bromodomain and extraterminal (BET) inhibitors), FMS-like tyrosine kinase receptor 3 (FLT3) inhibitors, and antibody-drug conjugates (vadastuximab), as well as cell cycle inhibitors (volasertib), B-cell lymphoma 2 (BCL-2) inhibitors, and aminopeptidase inhibitors. These agents are actively undergoing clinical investigation alone or in combination with available chemotherapy.
Collapse
Affiliation(s)
- Caner Saygin
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Hetty E. Carraway
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195 USA
- Department of Hematology and Oncology, Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Desk R30, Cleveland, OH 44195 USA
| |
Collapse
|
26
|
Boddu PC, Kantarjian HM, Ravandi F, Garcia-Manero G, Verstovsek S, Jabbour EJ, Takahashi K, Bhalla K, Konopleva M, DiNardo CD, Ohanian M, Pemmaraju N, Jain N, Pierce S, Wierda WG, Cortes JE, Kadia TM. Characteristics and outcomes of older patients with secondary acute myeloid leukemia according to treatment approach. Cancer 2017; 123:3050-3060. [PMID: 28387922 DOI: 10.1002/cncr.30704] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 02/20/2017] [Accepted: 03/07/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND The development of newer strategies to improve outcomes for older patients with secondary acute myeloid leukemia (s-AML) is a critical unmet need. Establishing baseline metrics for evaluating newer approaches is important. METHODS s-AML was defined as 1 or more of the following: a history of an antecedent hematologic disorder (AHD), a diagnosis of therapy-related acute myeloid leukemia (AML), and AML with karyotype abnormalities characteristic of myelodysplastic syndrome. Newly diagnosed s-AML patients aged 60 to 75 years were grouped into 5 treatment cohorts: 1) patients receiving high- or intermediate-dose cytarabine-based intensive chemotherapy (IC), 2) patients receiving a hypomethylating agent (HMA) or HMA combinations, 3) patients receiving low-dose cytarabine (LDAC) combinations, 4) patients receiving CPX-351, and 5) patients receiving investigational (INV) agents. Nine hundred thirty-one patients met the age and s-AML criteria. RESULTS Complete remission rates were statistically lower in the HMA group (36%) versus the IC (46%), CPX-351 (45%), and LDAC groups (43%). Patients receiving less intensive regimens (the HMA and LDAC groups combined) had superior overall survival (OS) in comparison with patients receiving IC-based regimens (median 6.9 vs 5.4 months; P = .048). Only 4.3% of the IC patients proceeded to transplantation, whereas 10.3% of the patients on lower intensity regimens did (P = .001). There was no difference in median survival between patients treated with CPX-351 and patients treated with conventional lower intensity approaches (P = .75). Age > 70 years, an adverse karyotype, and a prior AHD were associated with decreased OS in a multivariate analysis. CONCLUSIONS Lower intensity approaches are associated with lower early mortality rates and improved OS in comparison with intensive regimens. OS is poor with currently available therapies with a median OS of 6 months (5.4-7.6 months across regimens). Unsatisfactory outcomes with other INV agents underscore the need for more effective therapies. Cancer 2017;123:3050-60. © 2017 American Cancer Society.
Collapse
Affiliation(s)
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias J Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kapil Bhalla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maro Ohanian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sherry Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge E Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
27
|
Comparison of Reduced-Intensity Idarubicin and Daunorubicin Plus Cytarabine as Induction Chemotherapy for Elderly Patients with Newly Diagnosed Acute Myeloid Leukemia. Clin Drug Investig 2017; 37:167-174. [PMID: 27722823 DOI: 10.1007/s40261-016-0469-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVES The therapy in elderly patients with acute myeloid leukemia (AML) is a big challenge because of poor risk factors and inferior tolerance to intensive chemotherapy. This study aims to compare the efficacy between reduced-intensity idarubicin plus cytarabine and daunorubicin plus cytarabine (IA regimen and DA regimen, respectively) in elderly patients with newly diagnosed AML. METHODS We retrospectively investigated 74 patients with newly diagnosed non-M3 AML aged >60 years, where 33 patients received IA regimen, 30 patients received DA regimen, while 11 patients received supportive treatment. We observed the complete remission (CR) rates, overall survival (OS) and side effects in different arms. RESULTS The CR rate in IA arm (70.4 %, 19/27) was significantly higher than that in DA arm (40 %, 10/25) in de novo AML (p = 0.028), and further significantly higher when white blood cell (WBC) count >10 × 109/L (p = 0.042) and ECOG (Eastern Cooperative Oncology Group) score <2 (p = 0.021). The overall survival of the entire population was poor with a median survival of 10 months, 1- and 2-year survival rates were 40.5 % (30/74) and 9.5 % (7/74). The median survival of the patients with chemotherapy was 12 months, which was significantly longer than patients treated supportively (4 months) (p < 0.001). There were no differences of median survival and duration of CR between two arms. Early mortality decreased in the past 5 years in both groups. Meanwhile, low-dose idarubicin was well tolerated in elderly patients. CONCLUSIONS Reduced-intensity chemotherapy offered an improvement in survival, and the reduced-intensity IA regimen could improve CR rate in elderly patients with de novo AML.
Collapse
|
28
|
Podoltsev NA, Stahl M, Zeidan AM, Gore SD. Selecting initial treatment of acute myeloid leukaemia in older adults. Blood Rev 2016; 31:43-62. [PMID: 27745715 DOI: 10.1016/j.blre.2016.09.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 08/05/2016] [Accepted: 09/30/2016] [Indexed: 11/30/2022]
Abstract
More than half of the patients with acute myeloid leukaemia (AML) are older than 60years. The treatment outcomes in this group remain poor with a median overall survival of <1year. Selecting initial treatment for these patients involves an assessment of 'fitness' for induction chemotherapy. This is done based on patient and disease-related characteristics which help to estimate treatment-related mortality and chance of complete remission with induction chemotherapy. If the risk of treatment-related mortality is high and/or the likelihood of a patient achieving a complete remission is low, lower-intensity treatment (low-dose cytarabine, decitabine and azacitidine) should be discussed. As outcomes in both groups of patients remain poor, enrolment into clinical trials of novel agents with varying mechanisms of action should be considered for all older adults with AML. Novel agents in Phase III development include CPX-351, guadecitabine (SGI-110), quizartinib, crenolanib, sapacitabine, vosaroxin and volasertib.
Collapse
Affiliation(s)
- Nikolai A Podoltsev
- Department of Internal Medicine, Hematology Section, Yale University School of Medicine, New Haven, CT, USA.
| | - Maximilian Stahl
- Yale Traditional Internal Medicine Program, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Amer M Zeidan
- Department of Internal Medicine, Hematology Section, Yale University School of Medicine, New Haven, CT, USA.
| | - Steven D Gore
- Department of Internal Medicine, Hematology Section, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
29
|
Schliemann C, Gerss J, Wiebe S, Mikesch JH, Knoblauch N, Sauer T, Angenendt L, Kewitz T, Urban M, Butterfass-Bahloul T, Edemir S, Vehring K, Müller-Tidow C, Berdel WE, Krug U. A Phase I Dose Escalation Study of the Triple Angiokinase Inhibitor Nintedanib Combined with Low-Dose Cytarabine in Elderly Patients with Acute Myeloid Leukemia. PLoS One 2016; 11:e0164499. [PMID: 27716819 PMCID: PMC5055288 DOI: 10.1371/journal.pone.0164499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/21/2016] [Indexed: 11/18/2022] Open
Abstract
Nintedanib (BIBF 1120), a potent multikinase inhibitor of VEGFR-1/-2/-3, FGFR-1/-2/-3 and PDGFR-α/-β, exerts growth inhibitory and pro-apoptotic effects in myeloid leukemic cells, especially when used in combination with cytarabine. This phase I study evaluated nintedanib in combination with low-dose cytarabine (LDAC) in elderly patients with untreated or relapsed/refractory acute myeloid leukemia (AML) ineligible for intensive chemotherapy in a 3+3 design. Nintedanib (dose levels 100, 150, and 200 mg orally twice daily) and LDAC (20 mg subcutaneous injection twice daily for 10 days) were administered in 28-day cycles. Dose-limiting toxicity (DLT) was defined as non-hematological severe adverse reaction CTC grade ≥ 4 with possible or definite relationship to nintedanib. Between April 2012 and October 2013, 13 patients (median age 73 [range: 62-86] years) were enrolled. One patient did not receive study medication and was replaced. Nine (69%) patients had relapsed or refractory disease and 6 (46%) patients had unfavorable cytogenetics. The most frequently reported treatment-related adverse events (AE) were gastrointestinal events. Twelve SAEs irrespective of relatedness were reported. Two SUSARs were observed, one fatal hypercalcemia and one fatal gastrointestinal infection. Two patients (17%) with relapsed AML achieved a complete remission (one CR, one CRi) and bone marrow blast reductions without fulfilling PR criteria were observed in 3 patients (25%). One-year overall survival was 33%. Nintedanib combined with LDAC shows an adequate safety profile and survival data are promising in a difficult-to-treat patient population. Continuation of this trial with a phase II recommended dose of 2 x 200 mg nintedanib in a randomized, placebo-controlled phase II study is planned. The trial is registered to EudraCT as 2011-001086-41. TRIAL REGISTRATION ClinicalTrials.gov NCT01488344.
Collapse
Affiliation(s)
| | - Joachim Gerss
- Institute of Biostatistics and Clinical Research, University Hospital Muenster, Muenster, Germany
| | - Stefanie Wiebe
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Nicola Knoblauch
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Tim Sauer
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Linus Angenendt
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Tobias Kewitz
- Centre for Clinical Trials, University Hospital Muenster, Muenster, Germany
| | - Marc Urban
- Centre for Clinical Trials, University Hospital Muenster, Muenster, Germany
| | | | - Sabine Edemir
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Kerstin Vehring
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | | | - Wolfgang E. Berdel
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
| | - Utz Krug
- Department of Medicine A, University Hospital Muenster, Muenster, Germany
- * E-mail:
| |
Collapse
|
30
|
Benton CB, Ravandi F. Targeting acute myeloid leukemia with TP53-independent vosaroxin. Future Oncol 2016; 13:125-133. [PMID: 27615555 DOI: 10.2217/fon-2016-0300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Vosaroxin is a quinolone compound that intercalates DNA and induces TP53-independent apoptosis, demonstrating activity against acute myeloid leukemia (AML) in Phase I-III trials. Here, we examine vosaroxin's mechanism of action and pharmacology, and we review its use in AML to date, focusing on details of individual clinical trials. Most recently, when combined with cytarabine in a randomized Phase III trial (VALOR), vosaroxin improved outcomes versus cytarabine alone for relapsed/refractory AML in patients older than 60 years and for patients in early relapse. We consider its continued role in the context of a multifaceted strategy against AML, including its current use in clinical trials. Prospective use will define its role in the evolving landscape of AML therapy.
Collapse
Affiliation(s)
- Christopher B Benton
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
31
|
Marx KR, Kantarjian H, Ravandi F. Vosaroxin: innovative anticancer quinolone for the treatment of acute myelogenous leukemia. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1194753] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
32
|
Sanford D, Ravandi F. Management of Newly Diagnosed Acute Myeloid Leukemia in the Elderly: Current Strategies and Future Directions. Drugs Aging 2016; 32:983-97. [PMID: 26446152 DOI: 10.1007/s40266-015-0309-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The incidence of acute myeloid leukemia (AML) increases with age, and the majority of cases occur in adults aged >55 years. The prognosis of AML in older adults is generally poor; however, AML is a heterogeneous disease regardless of age, and prognosis depends on cytogenetic changes, genetic mutations, and patient characteristics. Several lines of evidence support offering treatment to the vast majority of older patients, and the survival benefit associated with this approach generally outweighs the risk of toxicity. Response and long-term survival using intensive induction regimens are significantly lower in older patients, although a small proportion of patients can achieve durable remissions. Selection of patients for intensive induction therapy requires comprehensive assessment of disease characteristics, performance status, and comorbidities. In unfit patients, options for treatment include hypomethylating agents, low-dose ara-C, or consideration of a clinical trial if available. The incorporation of novel therapies into treatment, such as FLT3 inhibitors and antibody-drug conjugates, offers significant promise in older patients, although, thus far, increased responses using novel agents have often not translated to improved survival outcomes. The development of reduced-intensity conditioning regimens and improvements in supportive care has increased the use of allogeneic stem cell transplant (ASCT) in older patients. Selection of patients for ASCT requires an estimation of the trade-off between toxicity and risk of relapse.
Collapse
Affiliation(s)
- David Sanford
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Farhad Ravandi
- Department of Leukemia, Unit 428, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
33
|
Short NJ, Ravandi F. The safety and efficacy of vosaroxin in patients with first relapsed or refractory acute myeloid leukemia - a critical review. Expert Rev Hematol 2016; 9:529-34. [DOI: 10.1080/17474086.2016.1187063] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
34
|
Kadia TM, Ravandi F, Cortes J, Kantarjian H. New drugs in acute myeloid leukemia. Ann Oncol 2016; 27:770-8. [PMID: 26802152 PMCID: PMC4843183 DOI: 10.1093/annonc/mdw015] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 12/27/2022] Open
Abstract
The standard therapy for acute myeloid leukemia (AML) has not changed meaningfully for the past four decades. Improvements in supportive care and modifications to the dose and schedule of existing agents have led to steady improvements in outcomes. However, developing new therapies for AML has been challenging. Although there have been advances in understanding the biology of AML, translating this knowledge to viable treatments has been slow. Active research is currently ongoing to address this important need and several promising drug candidates are currently in the pipeline. Here, we review some of the most advanced and promising compounds that are currently in clinical trials and may have the potential to be part of our future armamentarium. These drug candidates range from cytotoxic chemotherapies, targeted small-molecule inhibitors, and monoclonal antibodies.
Collapse
Affiliation(s)
- T M Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, USA
| | - F Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Cortes
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, USA
| | - H Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
35
|
Sanz MA, Iacoboni G, Montesinos P, Venditti A. Emerging strategies for the treatment of older patients with acute myeloid leukemia. Ann Hematol 2016; 95:1583-93. [PMID: 27118541 DOI: 10.1007/s00277-016-2666-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 04/04/2016] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML) is the most common acute leukemia in adults, and its incidence increases with age. Clinical outcomes in younger patients have improved over the years but, unfortunately, there is little evidence for an equivalent improvement in outcome for older patients. Approximately 50 % of older patients who are able to receive intensive chemotherapy will achieve a complete remission; however, they face a much higher relapse rate than younger patients, and survival rates for this group are low. Therefore, there is an urgent need to improve outcomes in older patients with AML. In this article, we discuss current treatment paradigms for older patients with AML including the challenges faced when determining which patients are eligible for intensive chemotherapy. We then highlight new treatments in development that may benefit this patient group. Cytotoxic agents, hypomethylating agents, molecularly targeted agents, and cell cycle kinase inhibitors are discussed, with a focus on novel agents that have achieved an advanced stage of development. Overall, the treatment of AML in older patients remains a challenge and, whenever possible, treatment should be offered in the context of clinical trials and should be planned with curative intent.
Collapse
Affiliation(s)
- Miguel A Sanz
- Hematology Department, Valencia University Medical School, Hospital Universitari i Politècnic La Fe, Avinguda Fernando Abril Martorell, 106, Valencia, 46026, Spain. .,Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain.
| | - Gloria Iacoboni
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Pau Montesinos
- Department of Hematology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Adriano Venditti
- Department of Hematology, Tor Vergata University Hospital, Viale Oxford 81, 00133, Rome, Italy
| |
Collapse
|
36
|
Khwaja A, Bjorkholm M, Gale RE, Levine RL, Jordan CT, Ehninger G, Bloomfield CD, Estey E, Burnett A, Cornelissen JJ, Scheinberg DA, Bouscary D, Linch DC. Acute myeloid leukaemia. Nat Rev Dis Primers 2016; 2:16010. [PMID: 27159408 DOI: 10.1038/nrdp.2016.10] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukaemia (AML) is a disorder characterized by a clonal proliferation derived from primitive haematopoietic stem cells or progenitor cells. Abnormal differentiation of myeloid cells results in a high level of immature malignant cells and fewer differentiated red blood cells, platelets and white blood cells. The disease occurs at all ages, but predominantly occurs in older people (>60 years of age). AML typically presents with a rapid onset of symptoms that are attributable to bone marrow failure and may be fatal within weeks or months when left untreated. The genomic landscape of AML has been determined and genetic instability is infrequent with a relatively small number of driver mutations. Mutations in genes involved in epigenetic regulation are common and are early events in leukaemogenesis. The subclassification of AML has been dependent on the morphology and cytogenetics of blood and bone marrow cells, but specific mutational analysis is now being incorporated. Improvements in treatment in younger patients over the past 35 years has largely been due to dose escalation and better supportive care. Allogeneic haematopoietic stem cell transplantation may be used to consolidate remission in those patients who are deemed to be at high risk of relapse. A plethora of new agents - including those targeted at specific biochemical pathways and immunotherapeutic approaches - are now in trial based on improved understanding of disease pathophysiology. These advances provide good grounds for optimism, although mortality remains high especially in older patients.
Collapse
Affiliation(s)
- Asim Khwaja
- Department of Haematology, University College London, UCL Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Magnus Bjorkholm
- Department of Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Rosemary E Gale
- Department of Haematology, University College London, UCL Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Ross L Levine
- Human Oncology and Pathogenesis Program, Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Craig T Jordan
- Division of Hematology, University of Colorado Denver, Denver, Colorado, USA
| | - Gerhard Ehninger
- Department of Internal Medicine, Technical University Dresden, Dresden, Germany
| | | | - Eli Estey
- Division of Hematology, University of Washington and Clinical Research Division Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | | | | | - David A Scheinberg
- Molecular Pharmacology Program, Experimental Therapeutics Center, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Didier Bouscary
- Institut Cochin, Département Développement Reproduction Cancer, CNRS UMR8104, INSERM U1016, Paris, France.,Service d'Hématologie, Hôpital Cochin, AP-HP, Paris, France.,Université Paris Descartes, Faculté de Médecine Sorbonne Paris Cité, Paris, France
| | - David C Linch
- Department of Haematology, University College London, UCL Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
37
|
Amadori S, Suciu S, Selleslag D, Aversa F, Gaidano G, Musso M, Annino L, Venditti A, Voso MT, Mazzone C, Magro D, De Fabritiis P, Muus P, Alimena G, Mancini M, Hagemeijer A, Paoloni F, Vignetti M, Fazi P, Meert L, Ramadan SM, Willemze R, de Witte T, Baron F. Gemtuzumab Ozogamicin Versus Best Supportive Care in Older Patients With Newly Diagnosed Acute Myeloid Leukemia Unsuitable for Intensive Chemotherapy: Results of the Randomized Phase III EORTC-GIMEMA AML-19 Trial. J Clin Oncol 2016; 34:972-9. [PMID: 26811524 DOI: 10.1200/jco.2015.64.0060] [Citation(s) in RCA: 267] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To compare single-agent gemtuzumab ozogamicin (GO) with best supportive care (BSC) including hydroxyurea as first-line therapy in older patients with acute myeloid leukemia unsuitable for intensive chemotherapy. PATIENTS AND METHODS In this trial, patients at least 61 years old were centrally randomized (1:1) to receive either a single induction course of GO (6 mg/m(2) on day 1 and 3 mg/m(2) on day 8) or BSC. Patients who did not progress after GO induction could receive up to eight monthly infusions of the immunoconjugate at 2 mg/m(2). Randomization was stratified by age, WHO performance score, CD33 expression status, and center. The primary end point was overall survival (OS) by intention-to-treat analysis. RESULTS A total of 237 patients were randomly assigned (118 to GO and 119 to BSC). The median OS was 4.9 months (95% CI, 4.2 to 6.8 months) in the GO group and 3.6 months (95% CI, 2.6 to 4.2 months) in the BSC group (hazard ratio, 0.69; 95% CI, 0.53 to 0.90; P = .005); the 1-year OS rate was 24.3% with GO and 9.7% with BSC. The OS benefit with GO was consistent across most subgroups, and was especially apparent in patients with high CD33 expression status, in those with favorable/intermediate cytogenetic risk profile, and in women. Overall, complete remission (CR [complete remission] + CRi [CR with incomplete recovery of peripheral blood counts]) occurred in 30 of 111 (27%) GO recipients. The rates of serious adverse events (AEs) were similar in the two groups, and no excess mortality from AEs was observed with GO. CONCLUSION First-line monotherapy with low-dose GO, as compared with BSC, significantly improved OS in older patients with acute myeloid leukemia who were ineligible for intensive chemotherapy. No unexpected AEs were identified and toxicity was manageable.
Collapse
Affiliation(s)
- Sergio Amadori
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands.
| | - Stefan Suciu
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Dominik Selleslag
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Franco Aversa
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Gianluca Gaidano
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Maurizio Musso
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Luciana Annino
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Adriano Venditti
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Maria Teresa Voso
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Carla Mazzone
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Domenico Magro
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Paolo De Fabritiis
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Petra Muus
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Giuliana Alimena
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Marco Mancini
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Anne Hagemeijer
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Francesca Paoloni
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Marco Vignetti
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Paola Fazi
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Liv Meert
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Safaa Mahmoud Ramadan
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Roel Willemze
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Theo de Witte
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| | - Frédéric Baron
- Sergio Amadori, Adriano Venditti, and Maria Teresa Voso, Tor Vergata University; Luciana Annino, San Giovanni Addolorata Hospital; Paolo De Fabritiis, St Eugenio Hospital; Giuliana Alimena and Marco Mancini, Sapienza University; Francesca Paoloni, Marco Vignetti, and Paola Fazi, GIMEMA Foundation, Roma; Franco Aversa, University Hospital, Parma; Gainluca Gaidano, University of Eastern Piedmont, Novara; Maurizio Musso, La Maddalena Clinic, Palermo; Carla Mazzone, Annunziata Hospital, Cosenza; Domenico Magro, Pugliese-Ciaccio Hospital, Catanzaro; Safaa Mahmoud Ramadan, European Institute of Oncology, Milano, Italy; Stefan Suciu and Liv Meert, EORTC Headquarters, Brussels; Dominik Selleslag, AZ St Jan, Brugge; Anne Hagemeijer, KULeuven, Leuven; Frédéric Baron, Centre Hospitalier Universitaire, Liège, Belgium; Petra Muus and Theo de Witte, Radboudumc, Nijmegen; and Roel Willemze, University Medical Center, Leiden, the Netherlands
| |
Collapse
|
38
|
Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood 2016; 127:53-61. [PMID: 26660429 PMCID: PMC4705610 DOI: 10.1182/blood-2015-08-604520] [Citation(s) in RCA: 398] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/03/2015] [Indexed: 12/21/2022] Open
Abstract
Recent advances in acute myeloid leukemia (AML) biology and its genetic landscape should ultimately lead to more subset-specific AML therapies, ideally tailored to each patient's disease. Although a growing number of distinct AML subsets have been increasingly characterized, patient management has remained disappointingly uniform. If one excludes acute promyelocytic leukemia, current AML management still relies largely on intensive chemotherapy and allogeneic hematopoietic stem cell transplantation (HSCT), at least in younger patients who can tolerate such intensive treatments. Nevertheless, progress has been made, notably in terms of standard drug dose intensification and safer allogeneic HSCT procedures, allowing a larger proportion of patients to achieve durable remission. In addition, improved identification of patients at relatively low risk of relapse should limit their undue exposure to the risks of HSCT in first remission. The role of new effective agents, such as purine analogs or gemtuzumab ozogamicin, is still under investigation, whereas promising new targeted agents are under clinical development. In contrast, minimal advances have been made for patients unable to tolerate intensive treatment, mostly representing older patients. The availability of hypomethylating agents likely represents an encouraging first step for this latter population, and it is hoped will allow for more efficient combinations with novel agents.
Collapse
Affiliation(s)
- Hervé Dombret
- Department of Hematology, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France; Leukemia Translational Laboratory, EA3518, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France; and
| | - Claude Gardin
- Leukemia Translational Laboratory, EA3518, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France; and Department of Hematology, Hôpital Avicenne, AP-HP, Bobigny, France
| |
Collapse
|
39
|
Kantarjian H. Acute myeloid leukemia--major progress over four decades and glimpses into the future. Am J Hematol 2016; 91:131-45. [PMID: 26598393 DOI: 10.1002/ajh.24246] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022]
Abstract
In this Review, the progress in research and therapy of acute myeloid leukemia is detailed.
Collapse
Affiliation(s)
- Hagop Kantarjian
- Department of Leukemia; MD Anderson Cancer Center; Houston Texas
| |
Collapse
|
40
|
Abstract
Multiple new drugs are being developed to treat acute myeloid leukemia (AML), including novel formulations of traditional chemotherapy-antibody drug conjugates and agents that target specific mutant enzymes. Next-generation sequencing has allowed us to discover the genetic mutations that lead to the development and clinical progression of AML. Studies of clonal hierarchy suggest which mutations occur early and dominate. This has led to targeted therapy against mutant driver proteins as well as the development of drugs such as CPX-351 and SGN-CD33A whose mechanisms of action and efficacy may not be dependent on mutational complexity. In this brief review, we discuss drugs that may emerge as important for the treatment of AML in the next 10 years.
Collapse
|
41
|
Bornhäuser M. Vosaroxin in acute myeloid leukaemia. Lancet Oncol 2015; 16:1000-1001. [PMID: 26234173 DOI: 10.1016/s1470-2045(15)00165-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 07/14/2015] [Indexed: 11/18/2022]
|