51
|
Zhang Y, Archer KJ. Bayesian variable selection for high-dimensional data with an ordinal response: identifying genes associated with prognostic risk group in acute myeloid leukemia. BMC Bioinformatics 2021; 22:539. [PMID: 34727888 PMCID: PMC8565083 DOI: 10.1186/s12859-021-04432-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous cancer of the blood, though specific recurring cytogenetic abnormalities in AML are strongly associated with attaining complete response after induction chemotherapy, remission duration, and survival. Therefore recurring cytogenetic abnormalities have been used to segregate patients into favorable, intermediate, and adverse prognostic risk groups. However, it is unclear how expression of genes is associated with these prognostic risk groups. We postulate that expression of genes monotonically associated with these prognostic risk groups may yield important insights into leukemogenesis. Therefore, in this paper we propose penalized Bayesian ordinal response models to predict prognostic risk group using gene expression data. We consider a double exponential prior, a spike-and-slab normal prior, a spike-and-slab double exponential prior, and a regression-based approach with variable inclusion indicators for modeling our high-dimensional ordinal response, prognostic risk group, and identify genes through hypothesis tests using Bayes factor. RESULTS Gene expression was ascertained using Affymetrix HG-U133Plus2.0 GeneChips for 97 favorable, 259 intermediate, and 97 adverse risk AML patients. When applying our penalized Bayesian ordinal response models, genes identified for model inclusion were consistent among the four different models. Additionally, the genes included in the models were biologically plausible, as most have been previously associated with either AML or other types of cancer. CONCLUSION These findings demonstrate that our proposed penalized Bayesian ordinal response models are useful for performing variable selection for high-dimensional genomic data and have the potential to identify genes relevantly associated with an ordinal phenotype.
Collapse
Affiliation(s)
| | - Kellie J Archer
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
52
|
Awan UA, Farooq N, Sarwar A, Jehangir HMS, Hashmi MS, Alamgir M, Waheed F, Khurram M, Ahmed H, Khattak AA, Afzal MS. Cytogenetic abnormalities in patients with hematological malignancies in Lahore city, Pakistan. BRAZ J BIOL 2021; 83:e249911. [PMID: 34669802 DOI: 10.1590/1519-6984.249911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/03/2021] [Indexed: 11/22/2022] Open
Abstract
Hematological and hematopoietic cells malignancies of the genes and hematopoietic cells are associated with the genetic mutation, often at the chromosomal level. The standard cytogenetic study is widely accepted as one of the main diagnostics and prognostic determinants in patients. Therefore, the current descriptive and cross-sectional study sought to determine the cytogenetic analysis of frequent hematological malignancies in Pakistan. A total of 202 peripheral bone marrow or blood samples from patients with benign and malignant hematological malignancy were taken using a conventional G-banding technique. Among enrolled patients, the mean age was 21.5 years ± 23.4, and gender-wise distribution showed a marked predominance of the male 147 (73%) population compared to the female 55 (27%). Patients in the age group (2-10 years) had the highest frequency, 48 (24%), of hematological neoplasms, followed by age (11-20 years) with 40 (20%). Normal karyotypes (46, XX/46, XY) was found in 51% (n=103) patients. Furthermore, the frequency of complex karyotype was 30 (15%), while normal was seen in 171 (85%) patients. Pre-B Acute Lymphoblastic Leukemia (Pre-B ALL) was the most prevalent malignancy of 66 (33%), followed by Chronic Myelogenous Leukemia (CML) of 41 (20%) and Acute Lymphocytic Leukemia of 29 (14%). Translocation was the most prevalent 50 (25%), followed by hypotriploidy 14 (7%) and monosomy 8 (4%) on chromosome aberration analysis. In addition, t(9:22) translocation was found to be 20 (10%) in CML, with the majority in the age group (31-40 years). This study recommends that karyotyping should be tested frequently in hematological conditions because it may provide insight into the relative chromosomal changes associated with particular malignancies.
Collapse
Affiliation(s)
- U A Awan
- Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - N Farooq
- Lahore Medical and Dental College, Lahore, Pakistan
| | - A Sarwar
- Gujranwala Medical College, Gujranwala, Punjab, Pakistan
| | | | - M S Hashmi
- Bahria International Hospital, Lahore, Pakistan
| | - M Alamgir
- Punjab Institute of Cardiology, School of Allied Health Sciences, Lahore, Pakistan
| | - F Waheed
- Punjab University, Department of Microbiology and Molecular Genetics, Lahore, Pakistan
| | - M Khurram
- Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - H Ahmed
- COMSATS University, Department of Biosciences, Islamabad, Pakistan
| | - A A Khattak
- The University of Haripur, Department of Medical Laboratory Technology, Haripur, Khyber Pakhtunkhwa, Pakistan
| | - M S Afzal
- Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| |
Collapse
|
53
|
Lazarevic VL. Acute myeloid leukaemia in patients we judge as being older and/or unfit. J Intern Med 2021; 290:279-293. [PMID: 33780573 DOI: 10.1111/joim.13293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 11/29/2022]
Abstract
The definition of older age in AML is arbitrary. In the context of the clinical studies, it starts with age ≥60 or ≥65 years and in recent years ≥70 or 75, depending on the selection of the studied population. In clinical practice, with older age, we often mean that the patient is unfit for intensive chemotherapy. Higher age overlaps with categories such as worse performance status, unfitness, comorbidities, poor-risk cytogenetics, adverse mutation patterns, age-related clonal haematopoiesis and specific disease ontogeny. Intensive induction therapy can result in prolonged overall survival, at least in a subset of elderly patients aged up to 75 years despite the reluctance of some physicians and patients to use treatment regimens perceived as toxic. Venetoclax and azacitidine combination is the new standard of comparison for persons unfit for intensive therapy. New oral hypomethylating agent CC-486 as maintenance therapy led to a prolonged overall survival in a randomized trial of patients ≥55 years of age who were in first complete remission, but not eligible for allogeneic stem cell transplantation. Any therapy is better than no therapy, but a substantial proportion of older patients still receive only palliative care. Making a decision for AML diagnosed in older age should be individualized and shared through the dialog with the patient and relatives or cohabitants, considering medical issues and social factors including personal goals. Although we are witnesses of the advances in basic research and therapy, we are still a very long way from curing older patients with AML.
Collapse
Affiliation(s)
- V Lj Lazarevic
- From the, Department of Hematology, Oncology and Radiation Physics, Stem Cell Center, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
54
|
Kadia TM, Ravandi F, Borthakur G, Konopleva M, DiNardo CD, Daver N, Pemmaraju N, Kanagal-Shamanna R, Wang X, Huang X, Pierce S, Rausch C, Burger J, Ferrajoli A, Jain N, Popat U, Estrov Z, Verstovsek S, Jabbour E, Garcia-Manero G, Kantarjian H. Long-term results of low-intensity chemotherapy with clofarabine or cladribine combined with low-dose cytarabine alternating with decitabine in older patients with newly diagnosed acute myeloid leukemia. Am J Hematol 2021; 96:914-924. [PMID: 33901324 PMCID: PMC12002039 DOI: 10.1002/ajh.26206] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
The treatment of older patients with newly diagnosed acute myeloid leukemia (AML) using intensive chemotherapy is associated with treatment intolerance and poor survival. We evaluated two new lower-intensity regimens with clofarabine (n = 119) or cladribine (n = 129) combined with low-dose cytarabine (LDAC) alternating with decitabine. We reviewed response rates by subgroup and long term outcomes of 248 patients with newly diagnosed non core-binding-factor AML treated on two clinical trials investigating double nucleoside-analogue therapy (DNT) alternating with HMA from October, 2008 to April, 2018. Of 248 patients with a median age of 69 years (range, 49-85 years), 102 patients (41%) were ≥ 70 years, and 108 (44%) had adverse karyotype. Overall, 164 patients (66%) responded: 147 (59%) complete remission (CR) and 17 (7%) CR with incomplete count recovery (CRi). With a median follow up of 60 months, median relapse-free and overall survival (OS) were 10.8 and 12.5 months, respectively. The 2-year OS was 29%. Among patients with normal karyotype, the CR/CRi rate was 79% and the median OS 19.9 months. High response rates and OS were observed in patients with mutations in NPM1, FLT3, IDH2, and RUNX1. The 4- and 8-week mortality rates were 2% and 11%, respectively. The backbone of clofarabine or cladribine and LDAC alternating with decitabine was effective and safe for the treatment of older patients with newly diagnosed AML. Incorporating targeted therapies could extend the efficacy of this approach and provide more curative therapeutic options in this AML population.
Collapse
Affiliation(s)
- Tapan M. Kadia
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marina Konopleva
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D. DiNardo
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval Daver
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naveen Pemmaraju
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Xuemei Wang
- Departments of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xuelin Huang
- Departments of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sherry Pierce
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Caitlin Rausch
- Departments of Pharmacy, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jan Burger
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alessandra Ferrajoli
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nitin Jain
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Uday Popat
- Departments of Stem Cell Transplant, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zeev Estrov
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Srdan Verstovsek
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elias Jabbour
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Hagop Kantarjian
- Departments of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
55
|
McCurdy SR, Luger SM. Dose intensity for induction in acute myeloid leukemia: what, when, and for whom? Haematologica 2021; 106:2544-2554. [PMID: 34320781 PMCID: PMC8485660 DOI: 10.3324/haematol.2020.269134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Indexed: 11/09/2022] Open
Abstract
Intensive chemotherapy has been the backbone of the treatment of acute myeloid leukemia (AML) for decades. However, an increase in novel targeted agents, which has been brought about in part by a deeper understanding of the genetic makeup of AML, has led to remission-inducing regimens that do not require traditional cytotoxic agents. Combinations of a hypomethylating agent (HMA) and venetoclax have doubled the chance of remission for patients considered unfit for induction chemotherapy who would have traditionally been offered singleagent HMA. In fact, this regimen may rival the complete remission rate achieved with induction chemotherapy for certain populations such as the very elderly and those with secondary AML, but equivalency has yet to be established. Further advances include the addition of gemtuzumab ozogamicin and FLT3 inhibitors to induction chemotherapy, which improves survival for patients with core-binding factor and FLT3-mutated AML, respectively. Still, much work is needed to improve the outcomes of the highest-risk subgroups: frail patients and those with high-risk cytogenetics and/or TP53 mutations. Promisingly, the landscape of AML therapy is shifting dramatically and no longer is intensity, when feasible, always the best answer for AML.
Collapse
Affiliation(s)
- Shannon R McCurdy
- Division of Hematology-Oncology/Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Selina M Luger
- Division of Hematology-Oncology/Department of Medicine, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
56
|
Phase 3 randomized trial of chemotherapy with or without oblimersen in older AML patients: CALGB 10201 (Alliance). Blood Adv 2021; 5:2775-2787. [PMID: 34251414 DOI: 10.1182/bloodadvances.2021004233] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/08/2021] [Indexed: 11/20/2022] Open
Abstract
Overexpression of B-cell leukemia/lymphoma 2 (BCL2) renders acute myeloid leukemia (AML) cells resistant to chemotherapy and has been associated with unfavorable outcomes. Oblimersen (G3139) is a phosphorothioate 18-mer antisense oligonucleotide directed against the first 6 BCL2 codons. In a phase 1 study of AML patients treated with G3139, cytarabine, and daunorubicin induction with cytarabine consolidation, no antisense-related toxicity was reported, and BCL2 downregulation occurred in patients achieving complete remission. In this phase 3 trial, untreated older AML patients were randomized to cytarabine (100 mg/m2 per day on days 4-10) and daunorubicin (60 mg/m2 per day on days 4-6) followed by cytarabine consolidation (2000 mg/m2 per day on days 4-8) with (arm A) or without (arm B) G3139 (7 mg/m2 per day on days 1-10 [induction] or days 1-8 [consolidation]). A total of 506 patients were enrolled. No differences in toxicity were observed between arms. Estimated overall survival (OS) at 1 year was 43% for arm A and 40% for arm B (1-sided log rank P = .13), with no differences in disease-free (DFS; P = .26) or event-free survival (P = .80). Subgroup analyses showed patients age <70 years in arm A had improved OS by 1 month vs those in arm B (P = .04), and patients with secondary AML in arm A had better DFS vs those in arm B (P = .04). We conclude that addition of G3139 to chemotherapy failed to improve outcomes of older AML patients. However, more effective means of inhibiting BCL2 are showing promising results in combination with chemotherapy in AML. This trial was registered at www.clinicaltrials.gov as #NCT00085124.
Collapse
|
57
|
Bill M, Mrózek K, Giacopelli B, Kohlschmidt J, Nicolet D, Papaioannou D, Eisfeld AK, Kolitz JE, Powell BL, Carroll AJ, Stone RM, Garzon R, Byrd JC, Bloomfield CD, Oakes CC. Precision oncology in AML: validation of the prognostic value of the knowledge bank approach and suggestions for improvement. J Hematol Oncol 2021; 14:107. [PMID: 34229733 PMCID: PMC8261916 DOI: 10.1186/s13045-021-01118-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/25/2021] [Indexed: 12/18/2022] Open
Abstract
Recently, a novel knowledge bank (KB) approach to predict outcomes of individual patients with acute myeloid leukemia (AML) was developed using unbiased machine learning. To validate its prognostic value, we analyzed 1612 adults with de novo AML treated on Cancer and Leukemia Group B front-line trials who had pretreatment clinical, cytogenetics, and mutation data on 81 leukemia/cancer-associated genes available. We used receiver operating characteristic (ROC) curves and the area under the curve (AUC) to evaluate the predictive values of the KB algorithm and other risk classifications. The KB algorithm predicted 3-year overall survival (OS) probability in the entire patient cohort (AUCKB = 0.799), and both younger (< 60 years) (AUCKB = 0.747) and older patients (AUCKB = 0.770). The KB algorithm predicted non-remission death (AUCKB = 0.860) well but was less accurate in predicting relapse death (AUCKB = 0.695) and death in first complete remission (AUCKB = 0.603). The KB algorithm’s 3-year OS predictive value was higher than that of the 2017 European LeukemiaNet (ELN) classification (AUC2017ELN = 0.707, p < 0.001) and 2010 ELN classification (AUC2010ELN = 0.721, p < 0.001) but did not differ significantly from that of the 17-gene stemness score (AUC17-gene = 0.732, p = 0.10). Analysis of additional cytogenetic and molecular markers not included in the KB algorithm revealed that taking into account atypical complex karyotype, infrequent recurrent balanced chromosome rearrangements and mutational status of the SAMHD1, AXL and NOTCH1 genes may improve the KB algorithm. We conclude that the KB algorithm has a high predictive value that is higher than those of the 2017 and 2010 ELN classifications. Inclusion of additional genetic features might refine the KB algorithm.
Collapse
Affiliation(s)
- Marius Bill
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.
| | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA. .,The Ohio State Comprehensive Cancer Center, Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH, USA. .,The Ohio State University Comprehensive Cancer Center, 444 Tzagournis Medical Research Facility, 420 West 12th Avenue, Columbus, OH, 43210-1228, USA.
| | - Brian Giacopelli
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,The Ohio State Comprehensive Cancer Center, Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH, USA.,Alliance Statistics and Data Center, The Ohio State University Comprehensive, Cancer Center, Columbus, OH, USA
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,The Ohio State Comprehensive Cancer Center, Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH, USA.,Alliance Statistics and Data Center, The Ohio State University Comprehensive, Cancer Center, Columbus, OH, USA
| | - Dimitrios Papaioannou
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 400 West 12th Avenue, Wiseman Hall, Suite 455, Columbus, OH, 43210-1228, USA
| | - Ann-Kathrin Eisfeld
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,The Ohio State Comprehensive Cancer Center, Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 400 West 12th Avenue, Wiseman Hall, Suite 455, Columbus, OH, 43210-1228, USA
| | - Jonathan E Kolitz
- Zucker School of Medicine At Hofstra/Northwell, Northwell Health Cancer Institute, Lake Success, NY, USA
| | - Bayard L Powell
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | | | - Richard M Stone
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| | - Ramiro Garzon
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 400 West 12th Avenue, Wiseman Hall, Suite 455, Columbus, OH, 43210-1228, USA
| | - John C Byrd
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,The Ohio State Comprehensive Cancer Center, Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University, Columbus, OH, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 400 West 12th Avenue, Wiseman Hall, Suite 455, Columbus, OH, 43210-1228, USA
| | - Clara D Bloomfield
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA.,Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 400 West 12th Avenue, Wiseman Hall, Suite 455, Columbus, OH, 43210-1228, USA
| | - Christopher C Oakes
- The Ohio State University Comprehensive Cancer Center, 460 West 12th Avenue, Columbus, OH, 43210-1228, USA. .,Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, 400 West 12th Avenue, Wiseman Hall, Suite 455, Columbus, OH, 43210-1228, USA.
| |
Collapse
|
58
|
Phillips DF, Zeidner JF. Emerging therapies for AML with myelodysplasia-related changes: slowly but surely moving the needle. Expert Opin Emerg Drugs 2021; 26:245-257. [PMID: 34227451 DOI: 10.1080/14728214.2021.1950689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Patients with acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) have historically poor outcomes with conventional chemotherapy regimens. Current treatment strategies focus on intensive induction therapy followed by allogeneic stem cell transplant or a less intensive approach with hypomethylating agents with or without venetoclax. CPX-351 is a liposomal formulation of cytarabine and daunorubicin that has been shown to significantly improve response rates and survival compared with 7 + 3 (continuous infusion cytarabine plus anthracyclines). Despite the approval of CPX-351 for AML-MRC, overall prognosis remains poor with an unmet need to develop novel therapeutic strategies for this patient population.Areas covered: This article reviews the data for existing therapeutic options for patients with AML-MRC and the emerging therapies undergoing clinical trial development for this patient population.Expert opinion: The development of CPX-351 as a more effective induction therapeutic backbone for patients with AML-MRC presents an opportunity to investigate novel combination regimens in order to further improve outcomes. Promising emerging therapeutic modalities include immunotherapeutic strategies, small-molecule inhibitors and targeted agents. Unfortunately, there have been few clinical trials focusing on patients with AML-MRC with reliance instead on subgroup analyses. Clinical trials focused specifically on this patient population are urgently needed.
Collapse
Affiliation(s)
- Davis F Phillips
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Joshua F Zeidner
- University of North Carolina School of Medicine, Chapel Hill, NC, USA.,University of North Carolina School of Medicine, Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA.,Department of Medicine, Division of Hematology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
59
|
Vadakekolathu J, Minden MD, Hood T, Church SE, Reeder S, Altmann H, Sullivan AH, Viboch EJ, Patel T, Ibrahimova N, Warren SE, Arruda A, Liang Y, Smith TH, Foulds GA, Bailey MD, Gowen-MacDonald J, Muth J, Schmitz M, Cesano A, Pockley AG, Valk PJM, Löwenberg B, Bornhäuser M, Tasian SK, Rettig MP, Davidson-Moncada JK, DiPersio JF, Rutella S. Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci Transl Med 2021; 12:12/546/eaaz0463. [PMID: 32493790 DOI: 10.1126/scitranslmed.aaz0463] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/20/2020] [Accepted: 04/21/2020] [Indexed: 01/13/2023]
Abstract
Acute myeloid leukemia (AML) is a molecularly and clinically heterogeneous hematological malignancy. Although immunotherapy may be an attractive modality to exploit in patients with AML, the ability to predict the groups of patients and the types of cancer that will respond to immune targeting remains limited. This study dissected the complexity of the immune architecture of AML at high resolution and assessed its influence on therapeutic response. Using 442 primary bone marrow samples from three independent cohorts of children and adults with AML, we defined immune-infiltrated and immune-depleted disease classes and revealed critical differences in immune gene expression across age groups and molecular disease subtypes. Interferon (IFN)-γ-related mRNA profiles were predictive for both chemotherapy resistance and response of primary refractory/relapsed AML to flotetuzumab immunotherapy. Our compendium of microenvironmental gene and protein profiles provides insights into the immuno-biology of AML and could inform the delivery of personalized immunotherapies to IFN-γ-dominant AML subtypes.
Collapse
Affiliation(s)
| | - Mark D Minden
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - Tressa Hood
- NanoString Technologies Inc., Seattle, WA 98109, USA
| | | | - Stephen Reeder
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Heidi Altmann
- Department of Medicine, Universitätsklinikum Carl Gustav Carus, 01307 Dresden, Germany
| | | | | | - Tasleema Patel
- Department of Pediatrics, Division of Oncology and Centre for Childhood Cancer Research, Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, PA 19104, USA
| | - Narmin Ibrahimova
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | | | - Andrea Arruda
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - Yan Liang
- NanoString Technologies Inc., Seattle, WA 98109, USA
| | | | - Gemma A Foulds
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK
| | | | | | - John Muth
- MacroGenics Inc., Rockville, MD 20850, USA
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | | | - A Graham Pockley
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK.,Centre for Health, Ageing and Understanding Disease (CHAUD), Nottingham Trent University, Nottingham NG11 8NS, UK
| | - Peter J M Valk
- Department of Hematology, Erasmus University Medical Centre, 3000CA Rotterdam, Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus University Medical Centre, 3000CA Rotterdam, Netherlands
| | - Martin Bornhäuser
- Department of Medicine, Universitätsklinikum Carl Gustav Carus, 01307 Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sarah K Tasian
- Department of Pediatrics, Division of Oncology and Centre for Childhood Cancer Research, Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, PA 19104, USA
| | - Michael P Rettig
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | | | - John F DiPersio
- Division of Oncology, Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham NG11 8NS, UK. .,Centre for Health, Ageing and Understanding Disease (CHAUD), Nottingham Trent University, Nottingham NG11 8NS, UK
| |
Collapse
|
60
|
Fang H, Yabe M, Zhang X, Kim Y, Wu X, Wei P, Chi S, Zheng L, Garcia-Manero G, Shao L, Yuan J, Shen Y, Zheng G, Tang G, Wang W, Loghavi S, Shen Q, Yuan Y, He R, Chen D, Medeiros LJ, Hu S. Myelodysplastic syndrome with t(6;9)(p22;q34.1)/DEK-NUP214 better classified as acute myeloid leukemia? A multicenter study of 107 cases. Mod Pathol 2021; 34:1143-1152. [PMID: 33558656 DOI: 10.1038/s41379-021-00741-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 11/09/2022]
Abstract
t(6;9)(p22;q34.1)/DEK-NUP214 is a recurrent genetic abnormality that occurs in 1-2% of patients with acute myeloid leukemia (AML), and rarely in myelodysplastic syndrome (MDS). It has been suggested by others that all myeloid neoplasms with t(6;9)/DEK-NUP214 may be considered as AML, even when blast count is <20%. In this study, we compared the clinicopathologic features of 107 patients with myeloid neoplasms harboring t(6;9)/DEK-NUP214: 33 MDS and 74 AML. Compared with patients with AML, patients with MDS were older (p = 0.10), had a lower white blood cell count (p = 0.0017), a lower blast count in the peripheral blood (p < 0.0001) and bone marrow (p < 0.0001), a higher platelet count (p = 0.022), and a lower frequency of FLT3-ITD mutation (p = 0.01). In addition, basophilia was not a common feature in the patients of this cohort. Although there was no difference in overall survival between MDS and AML patients (p = 0.18) in the entire cohort, the survival curves did show a trend toward favorable survival in MDS patients. Multivariate analyses showed that initial diagnosis of MDS vs. AML and allogeneic hematopoietic stem cell transplantation were prognostic factors for survival of patients with t(6;9)/DEK-NUP214 (p = 0.008 and p < 0.0001, respectively). Our data suggest that MDS with t(6;9)/DEK-NUP214 is prognostically not equivalent to AML with t(6;9)/DEK-NUP214. These data also show that stem cell transplantation greatly improves the survival of MDS and AML patients with myeloid neoplasms associated with t(6;9)/DEK-NUP214.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Child
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomes, Human, Pair 6/genetics
- Chromosomes, Human, Pair 9/genetics
- Female
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Male
- Middle Aged
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/pathology
- Nuclear Pore Complex Proteins/genetics
- Oncogene Fusion
- Oncogene Proteins/genetics
- Oncogene Proteins, Fusion
- Poly-ADP-Ribose Binding Proteins/genetics
- Translocation, Genetic
- Young Adult
Collapse
Affiliation(s)
- Hong Fang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariko Yabe
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaohui Zhang
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Young Kim
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Xiaojun Wu
- Department of Pathology, John Hopkins University, Baltimore, MD, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunyi Chi
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Quantitative Sciences Program, The University of Texas MD Anderson Cancer Center/UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Lan Zheng
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Lina Shao
- Department of Pathology, The University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Ji Yuan
- Department of Pathology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yulei Shen
- Department of Pathology, The University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Gang Zheng
- Department of Pathology, John Hopkins University, Baltimore, MD, USA
| | - Guiling Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Shen
- Department of Pathology, Advent Health-Orlando, Orlando, FL, USA
| | - Yongzhong Yuan
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AK, USA
| | - Rong He
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Dong Chen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shimin Hu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
61
|
Secondary cytogenetic abnormalities in core-binding factor AML harboring inv(16) vs t(8;21). Blood Adv 2021; 5:2481-2489. [PMID: 34003250 DOI: 10.1182/bloodadvances.2020003605] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with core-binding factor (CBF) acute myeloid leukemia (AML), caused by either t(8;21)(q22;q22) or inv(16)(p13q22)/t(16;16)(p13;q22), have higher complete remission rates and longer survival than patients with other subtypes of AML. However, ∼40% of patients relapse, and the literature suggests that patients with inv(16) fare differently from those with t(8;21). We retrospectively analyzed 537 patients with CBF-AML, focusing on additional cytogenetic aberrations to examine their impact on clinical outcomes. Trisomies of chromosomes 8, 21, or 22 were significantly more common in patients with inv(16)/t(16;16): 16% vs 7%, 6% vs 0%, and 17% vs 0%, respectively. In contrast, del(9q) and loss of a sex chromosome were more frequent in patients with t(8;21): 15% vs 0.4% for del(9q), 37% vs 0% for loss of X in females, and 44% vs 5% for loss of Y in males. Hyperdiploidy was more frequent in patients with inv(16) (25% vs 9%, whereas hypodiploidy was more frequent in patients with t(8;21) (37% vs 3%. In multivariable analyses (adjusted for age, white blood counts at diagnosis, and KIT mutation status), trisomy 8 was associated with improved overall survival (OS) in inv(16), whereas the presence of other chromosomal abnormalities (not trisomy 8) was associated with decreased OS. In patients with t(8;21), hypodiploidy was associated with improved disease-free survival; hyperdiploidy and del(9q) were associated with improved OS. KIT mutation (either positive or not tested, compared with negative) conferred poor prognoses in univariate analysis only in patients with t(8;21).
Collapse
|
62
|
Combination of dasatinib with chemotherapy in previously untreated core binding factor acute myeloid leukemia: CALGB 10801. Blood Adv 2021; 4:696-705. [PMID: 32092139 DOI: 10.1182/bloodadvances.2019000492] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Acute myeloid leukemia (AML) with either t(8;21)(q22;q22) or inv(16)(p13q22)/t(16;16)(p13;q22) is referred to as core binding factor (CBF) AML. Although categorized as favorable risk, long-term survival for these patients is only ∼50% to 60%. Mutated (mut) or overexpressed KIT, a gene encoding a receptor tyrosine kinase, has been found almost exclusively in CBF AML and may increase the risk of disease relapse. We tested the safety and clinical activity of dasatinib, a multi-kinase inhibitor, in combination with chemotherapy. Sixty-one adult patients with AML and CBF fusion transcripts (RUNX1/RUNX1T1 or CBFB/MYH11) were enrolled on Cancer and Leukemia Group B (CALGB) 10801. Patients received cytarabine/daunorubicin induction on days 1 to 7 and oral dasatinib 100 mg/d on days 8 to 21. Upon achieving complete remission, patients received consolidation with high-dose cytarabine followed by dasatinib 100 mg/d on days 6 to 26 for 4 courses, followed by dasatinib 100 mg/d for 12 months. Fifteen (25%) patients were older (aged ≥60 years); 67% were CBFB/MYH11-positive, and 19% harbored KITmut. There were no unexpected or dose-limiting toxicities. Fifty-five (90%) patients achieved complete remission. With a median follow-up of 45 months, only 16% have relapsed. The 3-year disease-free survival and overall survival rates were 75% and 77% (79% and 85% for younger patients [aged <60 years], and 60% and 51% for older patients). Patients with KITmut had comparable outcome to those with wild-type KIT (3-year rates: disease-free survival, 67% vs 75%; overall survival, 73% vs 76%), thereby raising the question of whether dasatinib may overcome the negative impact of these genetic lesions. CALGB 10801 was registered at www.clinicaltrials.gov as #NCT01238211.
Collapse
|
63
|
Outcomes of older patients with NPM1-mutated AML: current treatments and the promise of venetoclax-based regimens. Blood Adv 2021; 4:1311-1320. [PMID: 32251497 DOI: 10.1182/bloodadvances.2019001267] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 02/19/2020] [Indexed: 12/13/2022] Open
Abstract
Nucleophosmin-1 mutations (NPM1+) occur in ∼30% of acute myeloid leukemia (AML) patients. Although typically associated with favorable prognosis, the beneficial impact of NPM1+ decreases with increasing age in patients treated with standard intensive chemotherapy (IC) or hypomethylating agents (HMAs). This retrospective analysis compared outcomes of NPM1+ AML patients treated with 1 of 3 induction approaches: HMA plus BCL-2 inhibitor venetoclax (VEN), HMA, or IC therapy. Composite complete response (CRc: CR + CR with incomplete count recovery) was seen in 96% (27/28), 36% (17/47), and 89% (204/228) of HMA + VEN, HMA, and IC patients, respectively (HMA + VEN vs HMA, P < .001; HMA + VEN vs IC, P = .10). Older patients (age >65 years) treated with HMA + VEN, HMA, or IC had CR rates of 88%, 28%, and 56%, respectively (HMA + VEN vs HMA, P < .001; HMA + VEN vs IC, P = .01). Significant improvement in overall survival (OS) was seen in patients age >65 years treated with HMA + VEN vs HMA (not reached [NR] vs 0.4 years; P < .001) or IC (NR vs 0.93 years; P = .001). Older patients treated with HMA + VEN had OS of 80% after median 1-year follow-up, with estimated 2-year OS of 70%. In the multivariable Cox model analysis, HMA + VEN was associated with a 69% lower risk of death compared with IC (hazard ratio, 0.31; 95% confidence interval, 0.12-0.83; type I error-adjusted P = .038). HMA + VEN combinations demonstrated impressive results compared with traditional standard-of-care regimens in older patients with NPM1+ AML.
Collapse
|
64
|
Prognostic impact of the ELN2017 risk classification in patients with AML receiving allogeneic transplantation. Blood Adv 2021; 4:3864-3874. [PMID: 32810221 DOI: 10.1182/bloodadvances.2020001904] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
In 2017, an updated European LeukemiaNet (ELN) risk classification was published allocating patients with acute myeloid leukemia (AML) to 3 risk groups on the basis of certain cytogenetic and molecular aberrations. To date, studies of the prognostic significance of the ELN2017 risk classification in the context of an allogeneic hematopoietic stem cell transplantation (HSCT) are lacking. We performed risk stratification according to the ELN2017 classification in 234 patients with AML who underwent allogeneic HSCT as a consolidation therapy. In our cohort, the risk of 39.7% of the patients was classified as favorable, that of 12.8% as intermediate, and that of 47.4% as adverse. In the context of allogeneic HSCT, the assignment to the 3 ELN2017 risk groups retained its prognostic significance, with patients with favorable risk having the best prognosis and those with adverse risk having the worst one. Subgroup analyses showed that patients with a monosomal karyotype or TP53 mutation had considerably increased relapse rates, even in the adverse-risk group. When we analyzed the impact of digital droplet PCR-based measurable residual disease (MRD) before allogeneic HSCT, MRD+ patients had impaired prognoses, with cumulative incidence of relapse and overall survival comparable to those of patients classified as having an ELN2017 adverse genetic risk. This study is the first to demonstrate that the ELN2017 classification distinguishes the 3 risk groups with significantly distinct prognoses, even after allogeneic HSCT, and emphasizes the dismal prognosis of patients with AML with TP53 mutations, monosomal karyotype, or MRD positivity after allogeneic HSCT.
Collapse
|
65
|
IDH2 mutations in patients with normal karyotype AML predict favorable responses to daunorubicin, cytarabine and cladribine regimen. Sci Rep 2021; 11:10017. [PMID: 33976256 PMCID: PMC8113255 DOI: 10.1038/s41598-021-88120-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
Mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2) genes occur in about 20% patients with acute myeloid leukemia (AML), leading to DNA hypermethylation and epigenetic deregulation. We assessed the prognostic significance of IDH1/2 mutations (IDH1/2+) in 398 AML patients with normal karyotype (NK-AML), treated with daunorubicine + cytarabine (DA), DA + cladribine (DAC), or DA + fludarabine. IDH2 mutation was an independent favorable prognostic factor for 4-year overall survival (OS) in total NK-AML population (p = 0.03, censoring at allotransplant). We next evaluated the effect of addition of cladribine to induction regimen on the patients’ outcome according to IDH1/2 mutation status. In DAC group, 4-year OS was increased in IDH2+ patients, compared to IDH-wild type group (54% vs 33%; p = 0.0087, censoring at allotransplant), while no difference was observed for DA-treated subjects. In multivariate analysis, DAC independently improved the survival of IDH2+ patients (HR = 0.6 [0.37–0.93]; p = 0.024; censored at transplant), indicating that this group specifically benefits from cladribine-containing therapy. In AML cells with R140Q or R172K IDH2 mutations, cladribine restrained mutations-related DNA hypermethylation. Altogether, DAC regimen produces better outcomes in IDH2+ NK-AML patients than DA, and this likely results from the hypomethylating activity of cladribine. Our observations warrant further investigations of induction protocols combining cladribine with IDH1/2 inhibitors in IDH2-mutant.
Collapse
|
66
|
Pugliese GM, Latini S, Massacci G, Perfetto L, Sacco F. Combining Mass Spectrometry-Based Phosphoproteomics with a Network-Based Approach to Reveal FLT3-Dependent Mechanisms of Chemoresistance. Proteomes 2021; 9:19. [PMID: 33925552 PMCID: PMC8167576 DOI: 10.3390/proteomes9020019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022] Open
Abstract
FLT3 mutations are the most frequently identified genetic alterations in acute myeloid leukemia (AML) and are associated with poor clinical outcome, relapse and chemotherapeutic resistance. Elucidating the molecular mechanisms underlying FLT3-dependent pathogenesis and drug resistance is a crucial goal of biomedical research. Given the complexity and intricacy of protein signaling networks, deciphering the molecular basis of FLT3-driven drug resistance requires a systems approach. Here we discuss how the recent advances in mass spectrometry (MS)-based (phospho) proteomics and multiparametric analysis accompanied by emerging computational approaches offer a platform to obtain and systematically analyze cell-specific signaling networks and to identify new potential therapeutic targets.
Collapse
Affiliation(s)
- Giusj Monia Pugliese
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (G.M.P.); (S.L.); (G.M.)
| | - Sara Latini
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (G.M.P.); (S.L.); (G.M.)
| | - Giorgia Massacci
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (G.M.P.); (S.L.); (G.M.)
| | - Livia Perfetto
- Fondazione Human Technopole, Department of Biology, Via Cristina Belgioioso 171, 20157 Milan, Italy;
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Via delle Ricerca Scientifica 1, 00133 Rome, Italy; (G.M.P.); (S.L.); (G.M.)
| |
Collapse
|
67
|
Vasconcelos FC, de Souza PS, Hancio T, de Faria FCC, Maia RC. Update on drug transporter proteins in acute myeloid leukemia: Pathological implication and clinical setting. Crit Rev Oncol Hematol 2021; 160:103281. [PMID: 33667660 DOI: 10.1016/j.critrevonc.2021.103281] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 12/11/2020] [Accepted: 02/27/2021] [Indexed: 12/17/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common hematological neoplasia causing death worldwide. The long-term overall survival is unsatisfactory due to many factors including older age, genetic heterogeneity and molecular characteristics comprising additional mutations, and resistance to chemotherapeutic drugs. The expression of ABCB1/P-glycoprotein, ABCC1/MRP1, ABCG2/BCRP and LRP transporter proteins is considered the major reason for multidrug resistance (MDR) in AML, however conflicting data have been reported. Here, we review the main issues about drug transporter proteins in AML clinical scenario, and highlight the clinicopathological significance of MDR phenotype associated with ABCB1 polymorphisms and FLT3 mutation.
Collapse
Affiliation(s)
- Flavia Cunha Vasconcelos
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
| | - Paloma Silva de Souza
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil; Laboratório de Produtos Bioativos, Polo Novo Cavaleiros/IMCT, Campus Professor Aloisio Teixeira (UFRJ/Macaé), Universidade Federal do Rio de Janeiro (UFRJ), Macaé, RJ, Brazil
| | - Thaís Hancio
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil; Programa de Pós-Graduação Stricto Sensu em Oncologia, INCA, RJ, Brazil
| | - Fernanda Costas Casal de Faria
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
| | - Raquel Ciuvalschi Maia
- Laboratório de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
68
|
Arslan S, Zhang J, Dhakal P, Moran J, Naidoo N, Lombardi J, Pullarkat V, Stein AS, Marcucci G, Yaghmour G, Bhatt VR, Fathi AT, Aldoss I. Outcomes of therapy with venetoclax combined with a hypomethylating agent in favorable-risk acute myeloid leukemia. Am J Hematol 2021; 96:E59-E63. [PMID: 33227142 DOI: 10.1002/ajh.26057] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Shukaib Arslan
- Department of Hematology and Hematopoietic Cell Transplantation City of Hope Duarte California USA
| | - Jianying Zhang
- Department of Information Sciences City of Hope Medical Center Duarte California USA
| | - Prajwal Dhakal
- Department of Hematology and Oncology University of Nebraska Medical Center Omaha Nebraska USA
| | - Jenna Moran
- Division of Hematology/Oncology Massachusetts General Hospital Boston Massachusetts USA
| | - Nuthana Naidoo
- Jane Anne Nohl Division of Hematology and Center for the study of Blood disease University of Southern California Los Angeles California USA
| | - Jennifer Lombardi
- Division of Hematology/Oncology Massachusetts General Hospital Boston Massachusetts USA
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation City of Hope Duarte California USA
| | - Anthony S. Stein
- Department of Hematology and Hematopoietic Cell Transplantation City of Hope Duarte California USA
| | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation City of Hope Duarte California USA
| | - George Yaghmour
- Jane Anne Nohl Division of Hematology and Center for the study of Blood disease University of Southern California Los Angeles California USA
| | - Vijaya R. Bhatt
- Department of Hematology and Oncology University of Nebraska Medical Center Omaha Nebraska USA
| | - Amir T. Fathi
- Division of Hematology/Oncology Massachusetts General Hospital Boston Massachusetts USA
| | - Ibrahim Aldoss
- Department of Hematology and Hematopoietic Cell Transplantation City of Hope Duarte California USA
| |
Collapse
|
69
|
Hansen DK, Kim J, Thompson Z, Hussaini M, Nishihori T, Ahmad A, Elmariah H, Faramand R, Mishra A, Davila ML, Khimani F, Lazaryan A, Sallman D, Liu H, Perez LE, Fernandez H, Nieder ML, Lancet JE, Pidala JA, Anasetti C, Bejanyan N. ELN 2017 Genetic Risk Stratification Predicts Survival of Acute Myeloid Leukemia Patients Receiving Allogeneic Hematopoietic Stem Cell Transplantation. Transplant Cell Ther 2021; 27:256.e1-256.e7. [PMID: 33781526 DOI: 10.1016/j.jtct.2020.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/11/2020] [Accepted: 12/19/2020] [Indexed: 12/17/2022]
Abstract
European LeukemiaNet (ELN) 2017 risk stratification by genetics is prognostic of outcomes in patients with acute myeloid leukemia (AML). However, the prognostic impact of the 2017 ELN genetic risk stratification after allogeneic hematopoietic cell transplantation (alloHCT) is not well established. We examined the effect of 2017 ELN genetic risk stratification on alloHCT outcomes of AML. We included 500 adult (≥18 years) AML patients in first (n = 370) or second (n = 130) complete remission receiving alloHCT from 2005 to 2016. Patients were classified into favorable (12%), intermediate (57%), and adverse (32%) 2017 ELN risk groups. The Cox proportional hazard model was used to conduct the multivariable analyses of leukemia-free survival (LFS) and overall survival (OS). Relapse and nonrelapse mortality were analyzed by the Fine-Gray regression model. OS at 2 years was 72% in the favorable versus 60% in the intermediate versus 45% in the adverse risk groups (P < .001). In multivariable analyses, the 2017 ELN classifier was an independent predictor of OS after alloHCT with significantly higher overall mortality in the intermediate (hazard ratio [HR] = 1.68, 95% confidence interval [CI], 1.06-2.68; P = .03) and adverse (HR = 2.50, 95% CI, 1.54-4.06; P < .001) risk groups compared to the favorable risk group. Similarly, LFS was worse in the intermediate (HR = 1.63, 95%, CI 1.06-2.53; P = .03) and adverse (HR 2.23, 95% CI, 1.41-3.54; P < .001) risk groups while relapse was higher in the adverse risk group (HR = 2.36, 95% CI, 1.28-4.35; P = .006) as compared to the favorable risk group. These data highlight the prognostic impact of the 2017 ELN genetic risk stratification on the survival of AML patients after alloHCT. Patients in the adverse risk group had the highest risk of relapse and worst survival. Thus the 2017 ELN prognostic system can help identify AML patients who may benefit from clinical trials offering relapse mitigation strategies to improve transplant outcomes.
Collapse
Affiliation(s)
- Doris K Hansen
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Zachary Thompson
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mohammad Hussaini
- Pathology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anam Ahmad
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hany Elmariah
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Rawan Faramand
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Asmita Mishra
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Marco L Davila
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Farhad Khimani
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aleksandr Lazaryan
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - David Sallman
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hien Liu
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Lia E Perez
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Hugo Fernandez
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Michael L Nieder
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jeffrey E Lancet
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joseph A Pidala
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Claudio Anasetti
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nelli Bejanyan
- Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
70
|
Tang H, Jia S, Bi L, Jia W, Gao G. Treatment options for older unfit patients with acute myeloid leukemia. Future Oncol 2021; 17:837-851. [PMID: 33522289 DOI: 10.2217/fon-2020-0615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Older acute myeloid leukemia patients usually experience a bleak outcome, especially those in the unfit group. For this unfit category, intensive chemotherapy and allogeneic stem cell transplantation are usually accompanied by higher early mortality, which results from higher risk genetic profiles and worse psychological and physiological conditions. The significant improvement in genetic technology recently has driven the appearance of several mutation-targeted therapies, such as FLT3, Bcl-2, IDH and Hedgehog pathway inhibitors and an anti-CD33 antibody-drug conjugate, which have changed enormously the therapeutic landscape of acute myeloid leukemia. This review describes the treatment dilemma of the unfit group and discusses the objective clinical data of each targeted drug and mechanisms of resistance, with a focus on combination strategies with fewer toxicities and abrogation of drug resistance.
Collapse
Affiliation(s)
- Hailong Tang
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Shuangshuang Jia
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Lei Bi
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Weijing Jia
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Guangxun Gao
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
71
|
Mitchell SR, Gopakumar J, Jaiswal S. Insights into clonal hematopoiesis and its relation to cancer risk. Curr Opin Genet Dev 2021; 66:63-69. [PMID: 33422951 DOI: 10.1016/j.gde.2020.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/23/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022]
Abstract
In the multi-hit model of carcinogenesis, a precancerous state often precedes overt malignancy. Identification of these states has been of great interest as they allow for early identification of at-risk individuals before the appearance of a future cancer. One such condition has recently been described for blood cancers: Clonal Hematopoiesis of Indeterminate Potential (CHIP). Recent research advances have elucidated the risk of progression of CHIP to myeloid malignancies, its potential as a precursor for non-myeloid blood cancers, and its association with non-hematological cancers. Understanding the evolution of CHIP to hematological malignancy may help identify CHIP carriers at high risk of transformation and lead to the development of targeted therapies that can be deployed preemptively.
Collapse
Affiliation(s)
- Shaneice R Mitchell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, United States
| | - Jayakrishnan Gopakumar
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, United States
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94304, United States; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94304, United States.
| |
Collapse
|
72
|
Goldsmith SR, Ghobadi A, DiPersio JF. Hematopoeitic Cell Transplantation and CAR T-Cell Therapy: Complements or Competitors? Front Oncol 2020; 10:608916. [PMID: 33415078 PMCID: PMC7783412 DOI: 10.3389/fonc.2020.608916] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/18/2020] [Indexed: 01/13/2023] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) and chimeric antigen receptor T cell (CAR T) therapy are the main modalities of adoptive cellular immunotherapy that have widely permeated the clinical space. The advent of both technologies revolutionized treatment of many hematologic malignancies, both offering the chance at sustained remissions for patients who would otherwise invariably succumb to their diseases. The understanding and exploitation of the nonspecific alloreactivity of allo-HCT and the graft-versus-tumor effect is contrasted by the genetically engineered precision of CAR T therapy. Historically, those with relapsed and refractory hematologic malignancies have often been considered for allo-HCT, although outcomes vary dramatically and are associated with potential acute and chronic toxicities. Such patients, mainly with B-lymphoid malignancies, may now be offered CAR T therapy. Yet, a lack of prospective data to guide decisions thereafter requires individualized approaches on whether to proceed to allo-HCT or observe. The continued innovations to make CAR T therapy more effective and accessible will continue to alter such approaches, but similar innovations in allo-HCT will likely result in similarly improved clinical outcomes. In this review, we describe the history of the two platforms, dissect the clinical indications emphasizing their intertwining and competitive roles described in trials and practice guidelines, and highlight innovations in which they complement or inform one another.
Collapse
Affiliation(s)
- Scott R. Goldsmith
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, United States
| | | | | |
Collapse
|
73
|
Chen EC, Garcia JS. Does patient fitness play a role in determining first-line treatment of acute myeloid leukemia? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:41-50. [PMID: 33275683 PMCID: PMC7727557 DOI: 10.1182/hematology.2020000087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The treatment choice for newly diagnosed patients with acute myeloid leukemia (AML) is no longer straightforward. Historically, patient fitness has been a major driver of the initial therapy decision based on the belief that intensive chemotherapy would be the optimal choice if a patient were "fit" enough to receive it. Tools based on chronological age, performance status, and comorbidities have been developed to help estimate patient fitness. With newer approved therapies that include nonintensive options such as IDH1 inhibition or less intensive options such as hypomethylating agent (HMA)- or low-dose cytarabine (LDAC)-based combinations with venetoclax, the choice of frontline AML therapy places more emphasis on disease-specific features, including cytogenetics and mutational profile. Moreover, newer treatments have higher response rates than what has been expected with older nonintensive options such as LDAC or HMA monotherapy. We present cases of three patients with AML with varying cytogenetic and molecular risks to demonstrate the important but changing role of patient fitness in the current era of expanding therapeutic options.
Collapse
Affiliation(s)
- Evan C Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | |
Collapse
|
74
|
Chimeric antigen receptor T cell therapies for acute myeloid leukemia. Front Med 2020; 14:701-710. [PMID: 33263835 DOI: 10.1007/s11684-020-0763-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/16/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor T cell (CAR T) therapies have achieved unprecedented efficacy in B-cell tumors, prompting scientists and doctors to exploit this strategy to treat other tumor types. Acute myeloid leukemia (AML) is a group of heterogeneous myeloid malignancies. Relapse remains the main cause of treatment failure, especially for patients with intermediate or high risk stratification. Allogeneic hematopoietic stem cell transplantation could be an effective therapy because of the graft-versus-leukemia effect, which unfortunately puts the patient at risk of serious complications, such as graft-versus-host disease. Although the identification of an ideal target antigen for AML is challenging, CAR T therapy remains a highly promising strategy for AML patients, particularly for those who are ineligible to receive a transplantation or have positive minimal residual disease. In this review, we focus on the most recent and promising advances in CAR T therapies for AML.
Collapse
|
75
|
Ihlow J, Gross S, Neuendorff NR, Busack L, Herneth A, Singh A, Schwarz M, Flörcken A, Anagnostopoulos I, Türkmen S, Burmeister T, Blau IW, Bullinger L, Westermann J. Clinical outcome of older adults with acute myeloid Leukemia: An analysis of a large tertiary referral Center over two decades. J Geriatr Oncol 2020; 12:540-549. [PMID: 33223482 DOI: 10.1016/j.jgo.2020.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/21/2020] [Accepted: 11/03/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE In older adults with acute myeloid leukemia (AML), the overall outcome is still dismal and long-term data on survival are scarce, particularly outside of clinical trials. Here, we assess characteristics, prognostic factors and long-term survival in patients ≥60 years who were treated for AML at our center over the past 17 years. METHODS 590 older adults with newly diagnosed AML were characterized according to Eastern Cooperative Oncology Group (ECOG) score, Charlson comorbidity index (CCI), European LeukemiaNet (ELN) risk, type of therapy, serum ferritin (SF) and further baseline characteristics. Survival analysis was performed accordingly. RESULTS Median age was 68 years and most patients were in good general condition. Median follow-up was 55.8 months. Of all patients, 66% received intensive chemotherapy (IC) +/- allogeneic hematopoietic stem cell transplantation (allo-HSCT). The remaining cohort received palliative chemotherapy (PC, 26%) or best supportive care only (BSC, 8%). Enrollment rate for interventional clinical trials was 26%. 5-year overall survival (OS) and relapse-free survival (RFS) were 18% (median 12.5 months) and 11,5% (median 10.0 months). Long-term survival was independently influenced by ECOG score, ELN risk group, baseline SF, previous myocardial infarction, and choice of therapy, but not consistently by age or CCI. Considering therapeutic subgroups, the contribution of particular parameters in predicting OS was most compelling in IC patients, but less consistent with PC or BSC. CONCLUSION Our results provide thorough insights into prognostication within therapeutic subgroups and emphasize the need for more detailed prognostic algorithms and routine geriatric assessment in the treatment of older adults with AML.
Collapse
Affiliation(s)
- Jana Ihlow
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| | - Sophia Gross
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Nina Rosa Neuendorff
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Leonie Busack
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Alma Herneth
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Anju Singh
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Michaela Schwarz
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Anne Flörcken
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Ioannis Anagnostopoulos
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Seval Türkmen
- Department of Medical Genetics and Human Genetics, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Burmeister
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Igor Wolfgang Blau
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Jörg Westermann
- Department of Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Berlin, Campus Virchow-Clinic, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| |
Collapse
|
76
|
Lomov N, Zerkalenkova E, Lebedeva S, Viushkov V, Rubtsov MA. Cytogenetic and molecular genetic methods for chromosomal translocations detection with reference to the KMT2A/MLL gene. Crit Rev Clin Lab Sci 2020; 58:180-206. [PMID: 33205680 DOI: 10.1080/10408363.2020.1844135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acute leukemias (ALs) are often associated with chromosomal translocations, in particular, KMT2A/MLL gene rearrangements. Identification or confirmation of these translocations is carried out by a number of genetic and molecular methods, some of which are routinely used in clinical practice, while others are primarily used for research purposes. In the clinic, these methods serve to clarify diagnoses and monitor the course of disease and therapy. On the other hand, the identification of new translocations and the confirmation of known translocations are of key importance in the study of disease mechanisms and further molecular classification. There are multiple methods for the detection of rearrangements that differ in their principle of operation, the type of problem being solved, and the cost-result ratio. This review is intended to help researchers and clinicians studying AL and related chromosomal translocations to navigate this variety of methods. All methods considered in the review are grouped by their principle of action and include karyotyping, fluorescence in situ hybridization (FISH) with probes for whole chromosomes or individual loci, PCR and reverse transcription-based methods, and high-throughput sequencing. Another characteristic of the described methods is the type of problem being solved. This can be the discovery of new rearrangements, the determination of unknown partner genes participating in the rearrangement, or the confirmation of the proposed rearrangement between the two genes. We consider the specifics of the application, the basic principle of each method, and its pros and cons. To illustrate the application, examples of studying the rearrangements of the KMT2A/MLL gene, one of the genes that are often rearranged in AL, are mentioned.
Collapse
Affiliation(s)
- Nikolai Lomov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Elena Zerkalenkova
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Svetlana Lebedeva
- Laboratory of Cytogenetics and Molecular Genetics Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Vladimir Viushkov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Mikhail A Rubtsov
- Department of Molecular Biology, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Department of Biochemistry, Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
77
|
Banskota SU, Khanal N, Bhatt VR. A precision medicine approach to management of acute myeloid leukemia in older adults. Curr Opin Oncol 2020; 32:650-655. [PMID: 32826488 PMCID: PMC7737662 DOI: 10.1097/cco.0000000000000673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
PURPOSE OF REVIEW Therapy selection in older adults with acute myeloid leukemia (AML) can be challenging because of a higher incidence of high-risk cytogenetic and molecular features conferring chemoresistance and poor functional status leading to increased treatment-related toxicities. The purpose of this review is to highlight the recent advances in precision medicine in AML that have shown promise to improve outcomes of older adults. RECENT FINDINGS The utilization of next generation sequencing to identify and target actionable mutations can influence therapy selection in one-third of patients and can result in higher response rates as well as survival compared with those who do not receive targeted therapy. Oral targeted agents are available for AML with IDH 1, IDH2, or FLT3 mutations. Low-intensity venetoclax-based regimens have shown high rates of responses in AML, particularly among those with NPM1 and IDH2 mutations; responses are often durable and associated with minimal residual disease (MRD) negativity. Multiple studies have demonstrated the prognostic significance of flow cytometric MRD, with potential implications for subsequent therapy. SUMMARY Novel approaches for AML risk-stratification, MRD assessment, and a precision medicine approach offer significant promise to improve survival and quality of life of older adults.
Collapse
Affiliation(s)
| | - Nabin Khanal
- Franciscan Physician Network Oncology & Hematology Specialists, St Francis hospital, Indianapolis, IN
| | - Vijaya Raj Bhatt
- Department of Internal Medicine, Division of Hematology-Oncology, University of Nebraska Medical Center, Omaha, NE
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
78
|
Mutational landscape and clinical outcome of patients with de novo acute myeloid leukemia and rearrangements involving 11q23/ KMT2A. Proc Natl Acad Sci U S A 2020; 117:26340-26346. [PMID: 33020282 DOI: 10.1073/pnas.2014732117] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Balanced rearrangements involving the KMT2A gene, located at 11q23, are among the most frequent chromosome aberrations in acute myeloid leukemia (AML). Because of numerous fusion partners, the mutational landscape and prognostic impact of specific 11q23/KMT2A rearrangements are not fully understood. We analyzed clinical features of 172 adults with AML and recurrent 11q23/KMT2A rearrangements, 141 of whom had outcome data available. We compared outcomes of these patients with outcomes of 1,097 patients without an 11q23/KMT2A rearrangement categorized according to the 2017 European LeukemiaNet (ELN) classification. Using targeted next-generation sequencing, we investigated the mutational status of 81 leukemia/cancer-associated genes in 96 patients with 11q23/KMT2A rearrangements with material for molecular studies available. Patients with 11q23/KMT2A rearrangements had a low number of additional gene mutations (median, 1; range 0 to 6), which involved the RAS pathway (KRAS, NRAS, and PTPN11) in 32% of patients. KRAS mutations occurred more often in patients with t(6;11)(q27;q23)/KMT2A-AFDN compared with patients with the other 11q23/KMT2A subsets. Specific gene mutations were too infrequent in patients with specific 11q23/KMT2A rearrangements to assess their associations with outcomes. We demonstrate that younger (age <60 y) patients with t(9;11)(p22;q23)/KMT2A-MLLT3 had better outcomes than patients with other 11q23/KMT2A rearrangements and those without 11q23/KMT2A rearrangements classified in the 2017 ELN intermediate-risk group. Conversely, outcomes of older patients (age ≥60 y) with t(9;11)(p22;q23) were poor and comparable to those of the ELN adverse-risk group patients. Our study shows that patients with an 11q23/KMT2A rearrangement have distinct mutational patterns and outcomes depending on the fusion partner.
Collapse
|
79
|
Zeidner JF, Lee DJ, Frattini M, Fine GD, Costas J, Kolibaba K, Anthony SP, Bearss D, Smith BD. Phase I Study of Alvocidib Followed by 7+3 (Cytarabine + Daunorubicin) in Newly Diagnosed Acute Myeloid Leukemia. Clin Cancer Res 2020; 27:60-69. [PMID: 32998965 DOI: 10.1158/1078-0432.ccr-20-2649] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/26/2020] [Accepted: 09/23/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Alvocidib is a cyclin-dependent kinase 9 inhibitor leading to downregulation of the antiapoptotic BCL-2 family member, MCL-1. Alvocidib has shown clinical activity in a timed sequential regimen with cytarabine and mitoxantrone in relapsed/refractory and newly diagnosed acute myeloid leukemia (AML) but has not been studied in combination with traditional 7+3 induction therapy. PATIENTS AND METHODS A multiinstitutional phase I dose-escalation study of alvocidib on days 1-3 followed by 7+3 (cytarabine 100 mg/m2/day i.v. infusion days 5-12 and daunorubicin 60 mg/m2 i.v. days 5-7) was performed in newly diagnosed AML ≤65 years. Core-binding factor AML was excluded. RESULTS There was no MTD on this study; the recommended phase II dose of alvocidib was 30 mg/m2 i.v. over 30 minutes followed by 60 mg/m2 i.v. infusion over 4 hours. There was one dose-limiting toxicity of cytokine release syndrome. The most common grade ≥3 nonhematologic toxicities were diarrhea (44%) and tumor lysis syndrome (34%). Overall, 69% (22/32) of patients achieved complete remission (CR). In an exploratory cohort, eight of nine (89%) patients in complete remission had no measurable residual disease, as determined by a centralized flow cytometric assay. Clinical activity was seen in patients with secondary AML, AML with myelodysplastic syndrome-related changes, and a genomic signature of secondary AML (50%, 50%, and 92% CR rates, respectively). CONCLUSIONS Alvocidib can be safely administered prior to 7+3 induction with encouraging clinical activity. These findings warrant further investigation of alvocidib combinations in newly diagnosed AML. This study was registered at clinicaltrials.gov identifier NCT03298984.
Collapse
Affiliation(s)
- Joshua F Zeidner
- University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina.
| | - Daniel J Lee
- Columbia University Medical Center, New York, New York
| | - Mark Frattini
- Columbia University Medical Center, New York, New York
- Celgene, Summit, New Jersey
| | - Gil D Fine
- Sumitomo Dainippon Pharma Oncology, Lehi, Utah
| | - Judy Costas
- Sumitomo Dainippon Pharma Oncology, Lehi, Utah
| | | | | | | | - B Douglas Smith
- Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| |
Collapse
|
80
|
Jain G, Thakral D, Sahoo RK, Kumar I, Vashishtha S, Verma P, Gupta R. Next generation sequencing guided treatment modulation and prognosis in Acute myeloid leukemia: Case vignettes. AMERICAN JOURNAL OF BLOOD RESEARCH 2020; 10:134-144. [PMID: 32923093 PMCID: PMC7486490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/05/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE The genomic mutational landscape of Acute Myeloid Leukemia has contributed to better treatment options, risk stratification and prognostication of this genetically heterogeneous disease. With several approved new drugs targeting specific mutations with better outcomes, we describe here two cases of AML in which, NPM1 was detected at diagnosis. The impact of age, type of treatment, stability of NPM1 mutation, and co-occurring mutations on survival are the essential parameters for investigation. METHOD Both the cases of AML were females, >60 years of age with normal 46XX karyotype. Allele specific RT-PCR and fragment analysis was performed for the detection of NPM1-A mutation at diagnosis. Both the patients were unfit for intensive chemotherapy therefore reduced intensity induction chemotherapy regimen was initially administered. Next-generation sequencing was performed for comprehensive mutational profiling, which guided targeted treatment, prognostic stratification, and response assessment. RESULT We report that the older AML patients with NPM1 mutation may not have a good outcome with intensive chemotherapy, especially patients with concurrent DNMT3A/IDH-1/2 mutations. In the second case with mutated NPM1, concurrent FLT3-ITD mutation served as a therapeutic target. The FLT3 inhibitor used in combination with standard therapy showed promising results in this case. CONCLUSION Here, we emphasize on the utility of next generation sequencing in guiding the treatment initiation or modulation during the disease course and risk stratification in AML. In conclusion, conventional chemotherapy in AML gives very poor overall survival rates and targeted chemotherapy against specific mutations may drastically improve patient survival and treatment outcomes.
Collapse
Affiliation(s)
- Garima Jain
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| | - Deepshi Thakral
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| | - Indresh Kumar
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| | - Sangeeta Vashishtha
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| | - Pramod Verma
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi, India
| |
Collapse
|
81
|
Abdallah M, Xie Z, Ready A, Manogna D, Mendler JH, Loh KP. Management of Acute Myeloid Leukemia (AML) in Older Patients. Curr Oncol Rep 2020; 22:103. [PMID: 32725515 DOI: 10.1007/s11912-020-00964-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW The acute myeloid leukemia (AML) treatment landscape has rapidly evolved over the past few years. These changes have several implications for the care of older adults (≥ 60 years), who have inferior clinical outcomes. We review decision-making in older adults, focusing on patient- and disease-related factors. We then summarize current treatment options, including multiple recently approved therapies, based on hypothetical clinical scenarios. RECENT FINDINGS In lieu of using chronological age to determine fitness, we highlight the importance of standardized fitness assessments using geriatric assessments. Next, we review intensive and lower-intensity treatment options in the upfront setting. We focus on multiple newly approved medications, including venetoclax, midostaurin, CPX-351, gemtuzumab, glasdegib, enasidenib, and ivosidenib, and their specific indications. Lastly, we briefly discuss supportive care of older adults with AML. Outcomes of older adults with AML remain poor; fortunately, there are many new promising treatment options. Personalized treatment plans based on patient- and disease-specific factors are essential to the care of older adults with AML.
Collapse
Affiliation(s)
- Maya Abdallah
- Sections of Hematology/Oncology & Geriatrics, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Zhuoer Xie
- Depatment of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Audrey Ready
- Department of Medicine, University of Massachusetts - Baystate Medical Center, Springfield, MA, USA
| | - Dharmini Manogna
- Department of Medicine, Rochester General Hospital, Rochester, NY, USA
| | - Jason H Mendler
- Division of Hematology/Oncology, University of Rochester Medical Center, James P. Wilmot Cancer Center, 601 Elmwood Avenue, Box 704, Rochester, NY, 14642, USA
| | - Kah Poh Loh
- Division of Hematology/Oncology, University of Rochester Medical Center, James P. Wilmot Cancer Center, 601 Elmwood Avenue, Box 704, Rochester, NY, 14642, USA.
| |
Collapse
|
82
|
Kirtonia A, Pandya G, Sethi G, Pandey AK, Das BC, Garg M. A comprehensive review of genetic alterations and molecular targeted therapies for the implementation of personalized medicine in acute myeloid leukemia. J Mol Med (Berl) 2020; 98:1069-1091. [PMID: 32620999 DOI: 10.1007/s00109-020-01944-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/18/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022]
Abstract
Acute myeloid leukemia (AML) is an extremely heterogeneous disease defined by the clonal growth of myeloblasts/promyelocytes not only in the bone marrow but also in peripheral blood and/or tissues. Gene mutations and chromosomal abnormalities are usually associated with aberrant proliferation and/or block in the normal differentiation of hematopoietic cells. So far, the combination of cytogenetic profiling and molecular and gene mutation analyses remains an essential tool for the classification, diagnosis, prognosis, and treatment for AML. This review gives an overview on how the development of novel innovative technologies has allowed us not only to detect the genetic alterations as early as possible but also to understand the molecular pathogenesis of AML to develop novel targeted therapies. We also discuss the remarkable advances made during the last decade to understand the AML genome both at primary and relapse diseases and how genetic alterations might influence the distinct biological groups as well as the clonal evolution of disease during the diagnosis and relapse. Also, the review focuses on how the persistence of epigenetic gene mutations during morphological remission is associated with relapse. It is suggested that along with the prognostic and therapeutic mutations, the novel molecular targeted therapies either approved by FDA or those under clinical trials including CART-cell therapy would be of immense importance in the effective management of AML.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology (AIB), Amity University, Gurgaon, Haryana, 122413, India
| | - Bhudev C Das
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
83
|
Minervini A, Coccaro N, Anelli L, Zagaria A, Specchia G, Albano F. HMGA Proteins in Hematological Malignancies. Cancers (Basel) 2020; 12:E1456. [PMID: 32503270 PMCID: PMC7353061 DOI: 10.3390/cancers12061456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/25/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
The high mobility group AT-Hook (HMGA) proteins are a family of nonhistone chromatin remodeling proteins known as "architectural transcriptional factors". By binding the minor groove of AT-rich DNA sequences, they interact with the transcription apparatus, altering the chromatin modeling and regulating gene expression by either enhancing or suppressing the binding of the more usual transcriptional activators and repressors, although they do not themselves have any transcriptional activity. Their involvement in both benign and malignant neoplasias is well-known and supported by a large volume of studies. In this review, we focus on the role of the HMGA proteins in hematological malignancies, exploring the mechanisms through which they enhance neoplastic transformation and how this knowledge could be exploited to devise tailored therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Francesco Albano
- Department of Emergency and Organ Transplantation (D.E.T.O.), Hematology Section, University of Bari, 70124 Bari, Italy; (A.M.); (N.C.); (L.A.); (A.Z.); (G.S.)
| |
Collapse
|
84
|
Stahl M, Tallman MS. Outcomes of allogeneic stem cell transplantation for patients with t(6:9) AML- A strong case for allogeneic stem cell transplantation in first complete remission. Br J Haematol 2020; 189:806-808. [PMID: 31999852 DOI: 10.1111/bjh.16478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 11/26/2022]
Affiliation(s)
- Maximilian Stahl
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin S Tallman
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
85
|
Eisfeld AK, Kohlschmidt J, Mims A, Nicolet D, Walker CJ, Blachly JS, Carroll AJ, Papaioannou D, Kolitz JE, Powell BE, Stone RM, de la Chapelle A, Byrd JC, Mrózek K, Bloomfield CD. Additional gene mutations may refine the 2017 European LeukemiaNet classification in adult patients with de novo acute myeloid leukemia aged <60 years. Leukemia 2020; 34:3215-3227. [PMID: 32461631 DOI: 10.1038/s41375-020-0872-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022]
Abstract
The European LeukemiaNet (ELN) recommendations for diagnosis and management of acute myeloid leukemia (AML) have become an important tool to assess patients' prognosis and guide treatment. We tested the prognostic impact of the 2017 ELN classification in a large cohort of 863 AML patients aged <60 years similarly treated on Cancer and Leukemia Group B/Alliance for Clinical Trials in Oncology studies. Based on multivariable models within each ELN genetic-risk group, we identified additional gene mutations that may refine the 2017 ELN risk classification. BCOR- or SETBP1-mutated favorable-risk patients with non-core-binding factor AML and IDH-mutated adverse-risk patients had intermediate-risk outcomes. Outcomes of NPM1/WT1 co-mutated patients and those of ZRSR2-mutated patients resembled outcome of adverse-risk patients. Moreover, FLT3-ITDhigh allelic ratio conferred adverse rather than intermediate-risk irrespective of the NPM1 mutation status, and DNMT3A mutations associated with very poor survival. Application of these refinements reclassified 9% of current favorable-risk patients and 53% of current intermediate-risk patients to the adverse-risk group, with similar poor survival as current adverse-risk patients. Furthermore, 4% of current favorable-risk patients and 9% of adverse-risk patients were reclassified to the intermediate-risk group.
Collapse
Affiliation(s)
- Ann-Kathrin Eisfeld
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.,Alliance Statistics and Data Center, The Ohio State University, Columbus, OH, USA
| | - Alice Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.,Alliance Statistics and Data Center, The Ohio State University, Columbus, OH, USA
| | | | - James S Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.,The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Dimitrios Papaioannou
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Jonathan E Kolitz
- Monter Cancer Center, Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA
| | - Bayard E Powell
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | | | | | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.,The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| | - Clara D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
86
|
Yanada M, Takami A, Yamasaki S, Arai Y, Konuma T, Uchida N, Najima Y, Fukuda T, Tanaka M, Ozawa Y, Ikegame K, Takanashi M, Ichinohe T, Okamoto S, Atsuta Y, Yano S. Allogeneic hematopoietic cell transplantation for adults with acute myeloid leukemia conducted in Japan during the past quarter century. Ann Hematol 2020; 99:1351-1360. [DOI: 10.1007/s00277-020-04051-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
|
87
|
Schönherz AA, Bødker JS, Schmitz A, Brøndum RF, Jakobsen LH, Roug AS, Severinsen MT, El-Galaly TC, Jensen P, Johnsen HE, Bøgsted M, Dybkær K. Normal myeloid progenitor cell subset-associated gene signatures for acute myeloid leukaemia subtyping with prognostic impact. PLoS One 2020; 15:e0229593. [PMID: 32324791 PMCID: PMC7179860 DOI: 10.1371/journal.pone.0229593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/10/2020] [Indexed: 12/30/2022] Open
Abstract
Acute myeloid leukaemia (AML) is characterised by phenotypic heterogeneity, which we hypothesise is a consequence of deregulated differentiation with transcriptional reminiscence of the normal compartment or cell-of-origin. Here, we propose a classification system based on normal myeloid progenitor cell subset-associated gene signatures (MAGS) for individual assignments of AML subtypes. We generated a MAGS classifier including the progenitor compartments CD34+/CD38- for haematopoietic stem cells (HSCs), CD34+/CD38+/CD45RA- for megakaryocyte-erythroid progenitors (MEPs), and CD34+/CD38+/CD45RA+ for granulocytic-monocytic progenitors (GMPs) using regularised multinomial regression with three discrete outcomes and an elastic net penalty. The regularisation parameters were chosen by cross-validation, and MAGS assignment accuracy was validated in an independent data set (N = 38; accuracy = 0.79) of sorted normal myeloid subpopulations. The prognostic value of MAGS assignment was studied in two clinical cohorts (TCGA: N = 171; GSE6891: N = 520) and had a significant prognostic impact. Furthermore, multivariate Cox regression analysis using the MAGS subtype, FAB subtype, cytogenetics, molecular genetics, and age as explanatory variables showed independent prognostic value. Molecular characterisation of subtypes by differential gene expression analysis, gene set enrichment analysis, and mutation patterns indicated reduced proliferation and overrepresentation of RUNX1 and IDH2 mutations in the HSC subtype; increased proliferation and overrepresentation of CEBPA mutations in the MEP subtype; and innate immune activation and overrepresentation of WT1 mutations in the GMP subtype. We present a differentiation-dependent classification system for AML subtypes with distinct pathogenetic and prognostic importance that can help identify candidates poorly responding to combination chemotherapy and potentially guide alternative treatments.
Collapse
Affiliation(s)
- Anna A. Schönherz
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Julie Støve Bødker
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Alexander Schmitz
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Rasmus Froberg Brøndum
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Lasse Hjort Jakobsen
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Anne Stidsholt Roug
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Marianne T. Severinsen
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Tarec C. El-Galaly
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Paw Jensen
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Hans Erik Johnsen
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Martin Bøgsted
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Clinical Medicine, Faculty of Medicine, Aalborg University, Aalborg, Denmark
- Department of Haematology, Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
- * E-mail:
| |
Collapse
|
88
|
Abstract
Introduction Although bibliometric analyses have been performed in the past on cancer and genomics, little is known about the most frequently cited articles specifically related to cancer epigenetics. Therefore, the purpose of this study is to use citation count to identify those papers in the scientific literature that have made key contributions in the field of cancer epigenetics and identify key driving forces behind future investigations. Materials and methods The Thomas Reuters Web of Science services was queried for the years 1980-2018 without language restrictions. Articles were sorted in descending order of the number of times they were cited in the Web of Science database by other studies, and all titles and abstracts were screened to identify the research areas of the top 100 articles. The number of citations per year was calculated. Results We identified the 100 most-cited articles on cancer epigenetics, which collectively had been cited 147,083 times at the time of this writing. The top-cited article was cited 7,124 times, with an average of 375 citations per year since publication. In the period 1980-2018, the most prolific years were the years 2006 and 2010, producing nine articles, respectively. Twenty-eight unique journals contributed to the 100 articles, with the Nature journal contributing most of the articles (n=22). The most common country of article origin was the United States of America (n=78), followed by Germany (n=4), Switzerland (n=4), Japan (n=3), Spain (n=2), and United Kingdom (n=2). Conclusions In this study, the 100 most-cited articles in cancer epigenetics were examined, and the contributions from various authors, specialties, and countries were identified. Cancer epigenetics is a rapidly growing scientific field impacting translational research in cancer screening, diagnosis, classification, prognosis, and targeted treatments. Recognition of important historical contributions to this field may guide future investigations.
Collapse
Affiliation(s)
- Ignacio Jusue-Torres
- Neurosurgery, Loyola University Chicago, Stritch School of Medicine, Chicago, USA
| | | | - Malcolm V Brock
- Surgery, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Alicia Hulbert
- Surgery, University of Illinois at Chicago, Chicago, USA
| |
Collapse
|
89
|
Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia. Leukemia 2020; 34:3161-3172. [PMID: 32231256 PMCID: PMC7685975 DOI: 10.1038/s41375-020-0806-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/10/2020] [Accepted: 03/13/2020] [Indexed: 12/17/2022]
Abstract
The revised 2017 European LeukemiaNet (ELN) recommendations for genetic risk stratification of acute myeloid leukemia have been widely adopted, but have not yet been validated in large cohorts of AML patients. We studied 1116 newly diagnosed AML patients (age range, 18–86 years) who had received induction chemotherapy. Among 771 patients not selected by genetics, the ELN-2017 classification re-assigned 26.5% of patients into a more favorable or, more commonly, a more adverse-risk group compared with the ELN-2010 recommendations. Forty percent of the cohort, and 51% of patients ≥60 years, were classified as adverse-risk by ELN-2017. In 599 patients <60 years, estimated 5-year overall survival (OS) was 64% for ELN-2017 favorable, 42% for intermediate-risk and 20% for adverse-risk patients. Among 517 patients aged ≥60 years, corresponding 5-year OS rates were 37, 16, and 6%. Patients with biallelic CEBPA mutations or inv(16) had particularly favorable outcomes, while patients with mutated TP53 and a complex karyotype had especially poor prognosis. DNMT3A mutations associated with inferior OS within each ELN-2017 risk group. Our results validate the prognostic significance of the revised ELN-2017 risk classification in AML patients receiving induction chemotherapy across a broad age range. Further refinement of the ELN-2017 risk classification is possible.
Collapse
|
90
|
Pan Y, Zhang Q, Deng X, An N, Du X, Liu J. Gene coexpression network analysis revealed biomarkers correlated with blast cells and survival in acute myeloid leukemia. Mol Clin Oncol 2020; 12:475-484. [PMID: 32257206 DOI: 10.3892/mco.2020.2006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy with a poorly understood pathogenesis, especially among patients with no known cytogenetic abnormalities. Furthermore, there is a lack of therapeutic gene targets and diagnostic biomarkers for the effective treatment of AML. The present study aimed to identify candidate biomarkers correlated with the clinical prognosis of patients with AML. Leukemic cells from 5 patients with AML exhibiting a normal karyotype, and hematopoietic cells from 5 healthy donors were processed for RNA sequencing (RNA-seq), and the obtained RNA expression profiles were subjected to weighted gene correlation network analysis. A novel group of genes (the red module) were identified to be significantly associated with AML, and this module contained a closely connected network with 147 nodes, which corresponded to 114 mRNAs. Analysis of the correlation between these mRNAs and blast cell percentage, overall survival (OS) and disease-free survival (DFS) using cases from The Cancer Genome Atlas (TCGA) database revealed that CSF3R, ALPL and LMTK2 were negatively associated with the percentage of blast cells, while high expression of these genes was associated with longer OS and DFS in patients with AML. The differential expression of these three genes between patients with AML and healthy control subjects was supported using the Genotype-Tissue Expression and TCGA databases and was further confirmed using reverse transcription-quantitative (RT-qPCR). These genes exhibited significantly lower expression in patients with AML compared with control subjects. The results indicated that CSF3R, ALPL and LMTK2 exhibit the potential to be prognostic biomarkers. However, the biological functions of these three candidate genes need to be assessed in further studies.
Collapse
Affiliation(s)
- Yuming Pan
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China.,Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Qiaoxia Zhang
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Xiaopeng Deng
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Na An
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Xin Du
- Shenzhen Bone Marrow Transplantation Public Service Platform, Department of Hematology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518035, P.R. China
| | - Jiajun Liu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
91
|
Lachowiez C, DiNardo CD, Konopleva M. Venetoclax in acute myeloid leukemia - current and future directions. Leuk Lymphoma 2020; 61:1313-1322. [PMID: 32031033 DOI: 10.1080/10428194.2020.1719098] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
B-cell leukemia/lymphoma-2 (BCL-2) inhibition with the targeted oral agent venetoclax (ABT-199) has reshaped the treatment landscape for multiple hematological malignancies. Venetoclax in combination with hypomethylating agents (HMAs) or low-dose cytarabine (LDAC) has led to improved outcomes in acute myeloid leukemia (AML) and represents a new standard of care for frontline AML treatment in older patients or those unfit for intensive chemotherapy. Combinations of venetoclax with standard induction therapy or targeted agents such as FLT-3 inhibitors and IDH inhibitors are leading to improved clinical outcomes, representing major advancements in a field that has been without significant changes in treatments for the last 30 years. This review provides biological and clinical rationale for current venetoclax based treatments in AML, addresses common adverse events encountered with venetoclax based therapy, and explores emerging clinical data regarding combinations of novel targeted therapeutics used in conjunction with venetoclax for the treatment of AML.
Collapse
Affiliation(s)
- Curtis Lachowiez
- Division of Cancer Medicine, M. D. Anderson Cancer Center, Houston, TX, USA
| | | | - Marina Konopleva
- Department of Leukemia, M. D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
92
|
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]
|
93
|
Brunner AM, Blonquist TM, DeAngelo DJ, McMasters M, Fell G, Hermance NM, Winer ES, Lindsley RC, Hobbs GS, Amrein PC, Hock HR, Steensma DP, Garcia JS, Luskin MR, Stone RM, Ballen KK, Rosenblatt J, Avigan D, Nahas MR, Mendez LM, McAfee SL, Moran JA, Bergeron M, Foster J, Bertoli C, Manning AL, McGregor KL, Fishman KM, Kuo FC, Baltay MT, Macrae M, Burke M, Behnan T, Wey MC, Som TT, Ramos AY, Rae J, Lombardi Story J, Nelson N, Logan E, Connolly C, Neuberg DS, Chen YB, Graubert TA, Fathi AT. Alisertib plus induction chemotherapy in previously untreated patients with high-risk, acute myeloid leukaemia: a single-arm, phase 2 trial. Lancet Haematol 2020; 7:e122-e133. [PMID: 31837959 PMCID: PMC10354959 DOI: 10.1016/s2352-3026(19)30203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/21/2019] [Accepted: 09/23/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Increased aurora A kinase (AAK) expression occurs in acute myeloid leukaemia; AAK inhibition is a promising therapeutic target in this disease. We therefore aimed to assess the activity of alisertib combined with 7 + 3 induction chemotherapy in previously untreated patients with high-risk acute myeloid leukaemia. METHODS We did a single-arm, phase 2 trial of patients recruited from the Dana-Farber/Harvard Cancer Center in the USA. Eligible patients had previously untreated acute myeloid leukaemia, an Eastern Cooperative Oncology Group performance status of 0-2, and were at high risk of disease as defined by the presence of an adverse-risk karyotype, the presence of secondary acute myeloid leukaemia arising from previous myelodysplastic syndrome or myeloproliferative neoplasm, the presence of therapy-related acute myeloid leukaemia, or being 65 years or older. Enrolled patients received 7 + 3 induction chemotherapy of continuous infusion of cytarabine (100 mg/m2 per day on days 1-7) and intravenous bolus of idarubicin (12 mg/m2 per day on days 1-3). Oral alisertib (30 mg) was given twice per day on days 8-15. Patients could receive up to four consolidation cycles with cytarabine and alisertib, and alisertib maintenance for 12 months. The primary endpoint was a composite including the proportion of patients achieving complete remission and those with a complete remission with incomplete neutrophil or platelet count recovery. Analyses were per-protocol. This study is registered with Clinicaltrials.gov, number NCT02560025, and has completed enrolment. FINDINGS Between Dec 31, 2015, and Aug 1, 2017, we enrolled a total of 39 eligible patients. 19 (49%) of 39 patients had secondary acute myeloid leukaemia and three (8%) had therapy-related acute myeloid leukaemia. At mid-induction, 33 (85%) of 39 patients showed marrow aplasia, six (15%) received re-induction. The median follow-up was 13·7 months (IQR 12·7-14·4). Composite remission was 64% (two-stage 95% CI 48-79), with 20 (51%) of 39 patients achieving complete remission and five (13%) achieving complete remission with incomplete neutrophil or platelet count recovery. The most common grade 3 or 4 adverse events included febrile neutropenia (16 [41%] of 39), neutropenia (12 [31%]), thrombocytopenia (13 [33%]), anaemia (11 [28%]), anorexia (nine [23%]), and oral mucositis (four [10%]). No treatment-related deaths were observed. INTERPRETATION These results suggest that alisertib combined with induction chemotherapy is active and safe in previously untreated patients with high-risk acute myeloid leukaemia. This study met criteria to move forward to a future randomised trial. FUNDING Millennium Pharmaceuticals.
Collapse
Affiliation(s)
- Andrew M Brunner
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Traci M Blonquist
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Daniel J DeAngelo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Geoffrey Fell
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nicole M Hermance
- Department of Biology, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Eric S Winer
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Gabriela S Hobbs
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Philip C Amrein
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Hanno R Hock
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - David P Steensma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Marlise R Luskin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Richard M Stone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Karen K Ballen
- Division of Hematology-Oncology, Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - David Avigan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Myrna R Nahas
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lourdes M Mendez
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Steven L McAfee
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jenna A Moran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Meghan Bergeron
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Julia Foster
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Christina Bertoli
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Amity L Manning
- Department of Biology, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Kristin L McGregor
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Kaitlyn M Fishman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Frank C Kuo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Michele T Baltay
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Molly Macrae
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Meghan Burke
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Tanya Behnan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Margaret C Wey
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Tina T Som
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Aura Y Ramos
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Jessica Rae
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Nicole Nelson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Emma Logan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christine Connolly
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Donna S Neuberg
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yi-Bin Chen
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Timothy A Graubert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Amir T Fathi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
94
|
Gianfaldoni G, Mannelli F, Intermesoli T, Bencini S, Giupponi D, Farina G, Cutini I, Bonetti MI, Masciulli A, Audisio E, Ferrero D, Pavoni C, Scattolin AM, Bosi A, Rambaldi A, Bassan R. Early peripheral clearance of leukemia-associated immunophenotypes in AML: centralized analysis of a randomized trial. Blood Adv 2020; 4:301-311. [PMID: 31978214 PMCID: PMC6988394 DOI: 10.1182/bloodadvances.2019000406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/20/2019] [Indexed: 12/15/2022] Open
Abstract
Although genetics is a relevant risk factor in acute myeloid leukemia (AML), it can be minimally informative and/or not readily available for the early identification of patients at risk for treatment failure. In a randomized trial comparing standard vs high-dose induction (ClinicalTrials.gov #NCT00495287), we studied early peripheral blast cell clearance (PBC) as a rapid predictive assay of chemotherapy response to determine whether it correlates with the achievement of complete remission (CR), as well as postremission outcome, according to induction intensity. Individual leukemia-associated immunophenotypes (LAIPs) identified pretherapy by flow cytometry were validated and quantified centrally after 3 days of treatment, expressing PBC on a logarithmic scale as the ratio of absolute LAIP+ cells on day 1 and day 4. Of 178 patients, 151 (84.8%) were evaluable. Patients in CR exhibited significantly higher median PBC (2.3 log) compared with chemoresistant patients (1.0 log; P < .0001). PBC < 1.0 predicted the worst outcome (CR, 28%). With 1.5 log established as the most accurate cutoff predicting CR, 87.5% of patients with PBC >1.5 (PBChigh, n = 96) and 43.6% of patients with PBC ≤1.5 (PBClow, n = 55) achieved CR after single-course induction (P < .0001). CR and PBChigh rates were increased in patients randomized to the high-dose induction arm (P = .04) and correlated strongly with genetic/cytogenetic risk. In multivariate analysis, PBC retained significant predictive power for CR, relapse risk, and survival. Thus, PBC analysis can provide a very early prediction of outcome, correlates with treatment intensity and disease subset, and may support studies of customized AML therapy.
Collapse
Affiliation(s)
- Giacomo Gianfaldoni
- Struttura Organizzativa Dipartimentale (SOD) Ematologia, Università di Firenze, and
| | - Francesco Mannelli
- Struttura Organizzativa Dipartimentale (SOD) Ematologia, Università di Firenze, and
- Centro Ricerca e Innovazione Malattie Mieloproliferative, Azienda Ospedaliero-Universitaria (AOU) Careggi, Florence, Italy
| | - Tamara Intermesoli
- Unità Strutturale Complessa di Ematologia, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Sara Bencini
- Struttura Organizzativa Dipartimentale (SOD) Ematologia, Università di Firenze, and
- Centro Diagnostico di Citofluorimetria e Immunoterapia, AOU Careggi, Florence, Italy
| | - Damiano Giupponi
- Unità Strutturale Complessa di Ematologia, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Giorgio Farina
- Unità Operativa Laboratorio Analisi-Citofluorimetria, Ospedale dell'Angelo, Mestre-Venezia, Italy
| | - Ilaria Cutini
- Struttura Organizzativa Dipartimentale (SOD) Ematologia, Università di Firenze, and
- SOD Terapie Cellulari e Medicina Trasfusionale, AOU Careggi, Florence, Italy
| | - Maria Ida Bonetti
- Struttura Organizzativa Dipartimentale (SOD) Ematologia, Università di Firenze, and
| | - Arianna Masciulli
- Unità Strutturale Complessa di Ematologia, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Ernesta Audisio
- Struttura Complessa di Ematologia, Ospedale Molinette, Turin, Italy
| | - Dario Ferrero
- Divisione di Ematologia, Università di Torino, Azienda Ospedaliera S. Giovanni Battista, Turin, Italy; and
| | - Chiara Pavoni
- Unità Strutturale Complessa di Ematologia, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | | | - Alberto Bosi
- Struttura Organizzativa Dipartimentale (SOD) Ematologia, Università di Firenze, and
| | - Alessandro Rambaldi
- Unità Strutturale Complessa di Ematologia, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Renato Bassan
- Unità Operativa di Ematologia, Ospedale dell'Angelo, Mestre-Venezia, Italy
| |
Collapse
|
95
|
Jiang H, Ou Z, He Y, Yu M, Wu S, Li G, Zhu J, Zhang R, Wang J, Zheng L, Zhang X, Hao W, He L, Gu X, Quan Q, Zhang E, Luo H, Wei W, Li Z, Zang G, Zhang C, Poon T, Zhang D, Ziyar I, Zhang RZ, Li O, Cheng L, Shimizu T, Cui X, Zhu JK, Sun X, Zhang K. DNA methylation markers in the diagnosis and prognosis of common leukemias. Signal Transduct Target Ther 2020; 5:3. [PMID: 32296024 PMCID: PMC6959291 DOI: 10.1038/s41392-019-0090-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/26/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022] Open
Abstract
The ability to identify a specific type of leukemia using minimally invasive biopsies holds great promise to improve the diagnosis, treatment selection, and prognosis prediction of patients. Using genome-wide methylation profiling and machine learning methods, we investigated the utility of CpG methylation status to differentiate blood from patients with acute lymphocytic leukemia (ALL) or acute myelogenous leukemia (AML) from normal blood. We established a CpG methylation panel that can distinguish ALL and AML blood from normal blood as well as ALL blood from AML blood with high sensitivity and specificity. We then developed a methylation-based survival classifier with 23 CpGs for ALL and 20 CpGs for AML that could successfully divide patients into high-risk and low-risk groups, with significant differences in clinical outcome in each leukemia type. Together, these findings demonstrate that methylation profiles can be highly sensitive and specific in the accurate diagnosis of ALL and AML, with implications for the prediction of prognosis and treatment selection.
Collapse
Affiliation(s)
- Hua Jiang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Zhiying Ou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yingyi He
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Meixing Yu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Shaoqing Wu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Gen Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jie Zhu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ru Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jiayi Wang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Lianghong Zheng
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Xiaohong Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenge Hao
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Liya He
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoqiong Gu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Qingli Quan
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Edward Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huiyan Luo
- State Key Laboratory of Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wei Wei
- State Key Laboratory of Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhihuan Li
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Guangxi Zang
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Charlotte Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Tina Poon
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Daniel Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ian Ziyar
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Run-Ze Zhang
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Oulan Li
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Linhai Cheng
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Taylor Shimizu
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| | - Xinping Cui
- Department of Statistics and Institute for Integrative Genome Biology, University of California Riverside, Riverside, CA, 92521, USA
| | - Jian-Kang Zhu
- Shanghai Center for Plant Stress Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai, 210602, China
| | - Xin Sun
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Kang Zhang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China.
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macau, China.
| |
Collapse
|
96
|
Rimando J, Slade M, DiPersio JF, Westervelt P, Gao F, Liu C, Romee R. The Predicted Indirectly Recognizable HLA Epitopes (PIRCHE) Score for HLA Class I Graft-versus-Host Disparity Is Associated with Increased Acute Graft-versus-Host Disease in Haploidentical Transplantation with Post-Transplantation Cyclophosphamide. Biol Blood Marrow Transplant 2020; 26:123-131. [PMID: 31563575 PMCID: PMC7286229 DOI: 10.1016/j.bbmt.2019.09.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 11/17/2022]
Abstract
The Predicted Indirectly Recognizable HLA Epitopes (PIRCHE) score quantifies the number of PIRCHEs in patient-donor pairs and represents an in silico measure of indirect alloreactivity. This biologic process is defined as T cell recognition of epitopes derived from mismatched, allogeneic HLA peptides that are subsequently presented by shared HLA molecules. Its association with clinical outcome has not been examined in haploidentical hematopoietic cell transplantation (haplo-HCT) with post-transplantation cyclophosphamide (PTCy). We hypothesized that the PIRCHE score (PS) would correlate with indirect alloreactivity and predict graft-versus-host disease (GVHD) risk and the incidence of relapse after haplo-HCT with PTCy. We retrospectively analyzed 148 patients who underwent peripheral blood stem cell T cell-replete haplo-HCT with PTCy at a single center between 2009 and 2016. For each patient-donor pair, the PS was calculated using the PIRCHE online matching tool. PSs were categorized by class and vector. The median class I graft-versus-host (GVH) PS was 11 (range, 0 to 56), and the median class I host-versus-graft (HVG) PS was 10 (range, 0 to 51). Class I GVH PS was associated with increased risk of grade II-IV acute GVHD (adjusted hazard ratio, 1.03 per PS unit increase; 95% confidence interval, 1.01 to 1.05; P= .008) but not of chronic GVHD or relapse. Our data show that use of the PS is a novel strategy for predicting clinical outcome in haplo-HCT; further studies using registry data and prospective cohorts are warranted to validate these findings.
Collapse
Affiliation(s)
- Joseph Rimando
- BMT and Leukemia Program, Washington University School of Medicine, Saint Louis, Missouri
| | - Michael Slade
- BMT and Leukemia Program, Washington University School of Medicine, Saint Louis, Missouri
| | - John F DiPersio
- BMT and Leukemia Program, Washington University School of Medicine, Saint Louis, Missouri
| | - Peter Westervelt
- BMT and Leukemia Program, Washington University School of Medicine, Saint Louis, Missouri
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Saint Louis, Missouri
| | - Chang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri.
| | - Rizwan Romee
- Division of Hematologic Malignancies and Transplantation, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
97
|
Pais A, Pande S, Pradhan G, Patil S. A complex karyotype with t(11;12)(q23;p13) translocation with coexistent clones of deletion 5q and cryptic deletion 7q in acute myeloid leukemia. Indian J Cancer 2020; 57:330-333. [DOI: 10.4103/ijc.ijc_473_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
98
|
Richardson DR, Foster MC, Coombs CC, Zeidner JF. Advances in Genomic Profiling and Risk Stratification in Acute Myeloid Leukemia. Semin Oncol Nurs 2019; 35:150957. [PMID: 31759819 PMCID: PMC10246438 DOI: 10.1016/j.soncn.2019.150957] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To review the current state of molecular and genetic profiling of acute myeloid leukemia (AML) and its implications. DATA SOURCE Peer-reviewed journal articles. CONCLUSION Significant advances in the understanding of the pathology of acute myeloid leukemia have led to refined risk stratification of patients and application of novel targeted therapies based on genetic profiles. Minimal residual disease testing allows for highly sensitive disease surveillance that can be used to predict relapse and assess treatment response. IMPLICATIONS FOR NURSING PRACTICE Accurate prognostication and therapeutic decision-making for patients with acute myeloid leukemia is dependent on molecular profiling. Being knowledgeable of the implications of minimal residual disease testing is critical for patient-centered care.
Collapse
Affiliation(s)
- Daniel R Richardson
- UNC Lineberger Comprehensive Cancer Center, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC; The Cecil G. Sheps Center for Health Services Research, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| | - Matthew C Foster
- UNC Lineberger Comprehensive Cancer Center, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Catherine C Coombs
- UNC Lineberger Comprehensive Cancer Center, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Joshua F Zeidner
- UNC Lineberger Comprehensive Cancer Center, Division of Hematology/Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
99
|
Tremblay G, Rousseau B, Marquis M, Beaubois C, Sauvageau G, Hébert J. Cost-Effectiveness Analysis of a HMGA2 Prognostic Test for Acute Myeloid Leukemia in a Canadian Setting. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2019; 17:827-839. [PMID: 31392669 PMCID: PMC6885508 DOI: 10.1007/s40258-019-00503-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND Current strategies for risk stratification of patients with acute myeloid leukemia assign approximately 40% of patients to the intermediate-risk group, where uncertainty about optimal therapy still persists. OBJECTIVE The objective of this study was to assess the cost effectiveness of a HMGA2 prognostic test based on HMGA2+/HMGA2- expression, which improves genetic risk stratification in acute myeloid leukemia, and compare this test with the current standard of care in Canada. METHODS A cost-effectiveness model was developed from the Canadian National Healthcare Service and societal perspective using data from the Quebec Leukemia Cell Bank, published literature, and physician surveys. The model includes a lifetime horizon assessing the HMGA2 test vs. standard of care. RESULTS The HMGA2 test outperformed the standard of care at all time horizons culminating with estimated improvements of 1.92 and 3.12 months in leukemia-free survival and overall survival, respectively. Costs associated with the HMGA2 test were consistently lower, except diagnostic costs, routine medical costs, and costs related to infections and false positives. From a societal perspective, total lifetime costs were $161,358 CAD and $151,908 CAD with the standard of care and the HMGA2 test, respectively. The incremental quality-adjusted life-year gain was 0.138, which led to dominance over the standard of care. Deterministic sensitivity analyses confirmed the results of the base-case scenario. Probabilistic sensitivity analyses revealed that for a willingness-to-pay threshold of $100,000 CAD, the probability of cost effectiveness was 87.19%. CONCLUSIONS The HMGA2 test is estimated to improve leukemia-free survival and overall survival outcomes, and yield costs savings from a healthcare system and societal perspective.
Collapse
Affiliation(s)
- Gabriel Tremblay
- Purple Squirrel Economics, New York, NY, USA.
- Geneconomics Inc, 1372 rue Du crépuscule, Lévis, QC, Canada, G7A 4K3.
| | | | - Miriam Marquis
- The Leucegene Project, Université de Montréal, Montreal, QC, Canada
- Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, 5415 l'Assomption Blvd, Montreal, QC, H1T 2M4, Canada
| | - Cyrielle Beaubois
- The Leucegene Project, Université de Montréal, Montreal, QC, Canada
- Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, 5415 l'Assomption Blvd, Montreal, QC, H1T 2M4, Canada
| | - Guy Sauvageau
- The Leucegene Project, Université de Montréal, Montreal, QC, Canada
- Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, 5415 l'Assomption Blvd, Montreal, QC, H1T 2M4, Canada
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Josée Hébert
- The Leucegene Project, Université de Montréal, Montreal, QC, Canada.
- Quebec Leukemia Cell Bank, Research Centre, Maisonneuve-Rosemont Hospital, 5415 l'Assomption Blvd, Montreal, QC, H1T 2M4, Canada.
- Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
100
|
Mendez LM, Posey RR, Pandolfi PP. The Interplay Between the Genetic and Immune Landscapes of AML: Mechanisms and Implications for Risk Stratification and Therapy. Front Oncol 2019; 9:1162. [PMID: 31781488 PMCID: PMC6856667 DOI: 10.3389/fonc.2019.01162] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022] Open
Abstract
AML holds a unique place in the history of immunotherapy by virtue of being among the first malignancies in which durable remissions were achieved with "adoptive immunotherapy," now known as allogeneic stem cell transplantation. The successful deployment of unselected adoptive cell therapy established AML as a disease responsive to immunomodulation. Classification systems for AML have been refined and expanded over the years in an effort to capture the variability of this heterogeneous disease and risk-stratify patients. Current systems increasingly incorporate information about cytogenetic alterations and genetic mutations. The advent of next generation sequencing technology has enabled the comprehensive identification of recurrent genetic mutations, many with predictive power. Recurrent genetic mutations found in AML have been intensely studied from a cell intrinsic perspective leading to the genesis of multiple, recently approved targeted therapies including IDH1/2-mutant inhibitors and FLT3-ITD/-TKD inhibitors. However, there is a paucity of data on the effects of these targeted agents on the leukemia microenvironment, including the immune system. Recently, the phenomenal success of checkpoint inhibitors and CAR-T cells has re-ignited interest in understanding the mechanisms leading to immune dysregulation and suppression in leukemia, with the objective of harnessing the power of the immune system via novel immunotherapeutics. A paradigm has emerged that places crosstalk with the immune system at the crux of any effective therapy. Ongoing research will reveal how AML genetics inform the composition of the immune microenvironment paving the way for personalized immunotherapy.
Collapse
Affiliation(s)
- Lourdes M. Mendez
- Department of Medicine and Pathology, Cancer Research Institute, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, United States
| | - Ryan R. Posey
- Department of Medicine and Pathology, Cancer Research Institute, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, United States
| | - Pier Paolo Pandolfi
- Department of Medicine and Pathology, Cancer Research Institute, Beth Israel Deaconess Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, United States
| |
Collapse
|