1
|
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: 56] [Impact Index Per Article: 14.0] [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
|
2
|
Clinical and molecular characterization of patients with acute myeloid leukemia and sole trisomies of chromosomes 4, 8, 11, 13 or 21. Leukemia 2019; 34:358-368. [PMID: 31462731 DOI: 10.1038/s41375-019-0560-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/01/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
Sole trisomies of chromosomes 4, 8, 11, 13 and 21 account for 89-95% of all sole trisomies in adult AML patients. We analyzed clinical and molecular characteristics of 138 de novo AML patients with sole +4, +8, +11, +13 or +21, and compared them with AML patients with those trisomies occurring in addition to other chromosome abnormalities (non-sole trisomy) and with cytogenetically normal AML (CN-AML) patients. Mutations in methylation-related genes were most commonly observed within each sole trisomy group (+4, 55%; +8, 58%; +11, 71%; +13, 71%; +21, 75% of patients). Patients with sole trisomies, excluding +4, also had frequent mutations in spliceosome genes (+8, 43%; +11, 65%; +13, 65%; +21, 45% of patients). In contrast, +4 patients frequently had mutations in transcription factor genes (44%) and NPM1 (36%). While 48% of patients with sole trisomies harbored mutations in a spliceosome gene, spliceosome mutations were observed in only 24% of non-sole trisomy (n = 131, P < 0.001) and 19% of CN-AML patients (n = 716, P < 0.001). Our data suggest that mutations affecting methylation-related genes are a molecular hallmark of sole trisomies. Mutations in spliceosome genes were also commonly observed in many sole trisomy patients and represent a novel finding in this cytogenetic subgroup.
Collapse
|
3
|
Bill M, Nicolet D, Kohlschmidt J, Walker CJ, Mrózek K, Eisfeld AK, Papaioannou D, Rong-Mullins X, Brannan Z, Kolitz JE, Powell BL, Archer KJ, Dorrance AM, Carroll AJ, Stone RM, Byrd JC, Garzon R, Bloomfield CD. Mutations associated with a 17-gene leukemia stem cell score and the score's prognostic relevance in the context of the European LeukemiaNet classification of acute myeloid leukemia. Haematologica 2019; 105:721-729. [PMID: 31413100 PMCID: PMC7049376 DOI: 10.3324/haematol.2019.225003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/13/2019] [Indexed: 02/06/2023] Open
Abstract
Leukemia stem cells (LSC) are more resistant to standard chemotherapy and their persistence during remission can cause relapse, which is still one of the major clinical challenges in the treatment of acute myeloid leukemia (AML). A better understanding of the mutational patterns and the prognostic impact of molecular markers associated with stemness could lead to better clinical management and improve patients’ outcomes. We applied a previously described 17-gene expression score comprising genes differently expressed between LSC and leukemic bulk blasts, for 934 adult patients with de novo AML, and studied associations of the 17-gene LSC score with clinical data and mutation status of 81 genes recurrently mutated in cancer and leukemia. We found that patients with a high 17-gene score were older and had more mutations. The 17-gene score was found to have a prognostic impact in both younger (aged <60 years) and older (aged ≥60 years) patients with AML. We also analyzed the 17-gene LSC score in the context of the 2017 European LeukemiaNet genetic-risk classification and found that for younger patients the score refined the classification, and identified patients currently classified in the European LeukemiaNet Favorable-risk category who had a worse outcome.
Collapse
Affiliation(s)
- Marius Bill
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.,Alliance Statistics and Data Center, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.,Alliance Statistics and Data Center, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | | | | | - Zachary Brannan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Jonathan E Kolitz
- Monter Cancer Center, Hofstra Northwell School of Medicine, Lake Success, NY
| | - Bayard L Powell
- Comprehensive Cancer Center of Wake Forest University, Winston-Salem, NC
| | - Kellie J Archer
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.,College of Public Health, The Ohio State University, Columbus, OH
| | - Adrienne M Dorrance
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John C Byrd
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Ramiro Garzon
- The Ohio State University Comprehensive Cancer Center, Columbus, OH.,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| | - Clara D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, OH .,Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
4
|
Walker CJ, Kohlschmidt J, Eisfeld AK, Mrózek K, Liyanarachchi S, Song C, Nicolet D, Blachly JS, Bill M, Papaioannou D, Oakes CC, Giacopelli B, Genutis LK, Maharry SE, Orwick S, Archer KJ, Powell BL, Kolitz JE, Uy GL, Wang ES, Carroll AJ, Stone RM, Byrd JC, de la Chapelle A, Bloomfield CD. Genetic Characterization and Prognostic Relevance of Acquired Uniparental Disomies in Cytogenetically Normal Acute Myeloid Leukemia. Clin Cancer Res 2019; 25:6524-6531. [PMID: 31375516 DOI: 10.1158/1078-0432.ccr-19-0725] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/06/2019] [Accepted: 07/30/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Uniparental disomy (UPD) is a way cancer cells duplicate a mutated gene, causing loss of heterozygosity (LOH). Patients with cytogenetically normal acute myeloid leukemia (CN-AML) do not have microscopically detectable chromosome abnormalities, but can harbor UPDs. We examined the prognostic significance of UPDs and frequency of LOH in patients with CN-AML.Experimental Design: We examined the frequency and prognostic significance of UPDs in a set of 425 adult patients with de novo CN-AML who were previously sequenced for 81 genes typically mutated in cancer. Associations of UPDs with outcome were analyzed in the 315 patients with CN-AML younger than 60 years. RESULTS We detected 127 UPDs in 109 patients. Most UPDs were large and typically encompassed all or most of the affected chromosome arm. The most common UPDs occurred on chromosome arms 13q (7.5% of patients), 6p (2.8%), and 11p (2.8%). Many UPDs significantly cooccurred with mutations in genes they encompassed, including 13q UPD with FLT3-internal tandem duplication (FLT3-ITD; P < 0.001), and 11p UPD with WT1 mutations (P = 0.02). Among patients younger than 60 years, UPD of 11p was associated with longer overall survival (OS) and 13q UPD with shorter disease-free survival (DFS) and OS. In multivariable models that accounted for known prognostic markers, including FLT3-ITD and WT1 mutations, UPD of 13q maintained association with shorter DFS, and UPD of 11p maintained association with longer OS. CONCLUSIONS LOH mediated by UPD is a recurrent feature of CN-AML. Detection of UPDs of 13q and 11p might be useful for genetic risk stratification of patients with CN-AML.
Collapse
Affiliation(s)
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Alliance Statistics and Data Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | - Chi Song
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, Ohio
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Alliance Statistics and Data Center, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - James S Blachly
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Marius Bill
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | - Brian Giacopelli
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Luke K Genutis
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Sophia E Maharry
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Shelley Orwick
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Kellie J Archer
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, Ohio
| | - Bayard L Powell
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina
| | - Jonathan E Kolitz
- Monter Cancer Center, Zucker School of Medicine at Hofstra/Northwell, Lake Success, New York
| | - Geoffrey L Uy
- Washington University School of Medicine in St. Louis, Siteman Cancer Center, St. Louis, Missouri
| | - Eunice S Wang
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | | | - John C Byrd
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | |
Collapse
|
5
|
Mrózek K, Eisfeld AK, Kohlschmidt J, Carroll AJ, Walker CJ, Nicolet D, Blachly JS, Bill M, Papaioannou D, Wang ES, Uy GL, Kolitz JE, Powell BL, Blum W, Stone RM, Byrd JC, Bloomfield CD. Complex karyotype in de novo acute myeloid leukemia: typical and atypical subtypes differ molecularly and clinically. Leukemia 2019; 33:1620-1634. [PMID: 30737482 PMCID: PMC6609457 DOI: 10.1038/s41375-019-0390-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/17/2018] [Accepted: 01/09/2019] [Indexed: 02/04/2023]
Abstract
Complex karyotype (CK) with ≥ 3 abnormalities is detected in 10-12% of patients with acute myeloid leukemia (AML) and associated with poor prognosis. The most common unbalanced abnormalities found in CK result in loss of material from the 5q, 7q, and/or 17p chromosome arms. The presence of 5q, 7q, and/or 17p abnormalities denotes typical CK and their absence denotes atypical CK. Since molecular features of CK-AML are not well characterized, we investigated mutational status of 81 leukemia/cancer-associated genes in 160 clinically well-characterized patients. They included 136 patients with ≥ 3 exclusively unbalanced chromosome abnormalities, 96 of whom had a typical CK and 40 atypical CK, and 24 patients with ≥ 1 balanced abnormality in addition to ≥ 2 unbalanced ones. Patients with atypical CK-AML differed from those with typical CK-AML: they carried TP53 mutations less often (P < 0.001) and more often PHF6 (P = 0.008), FLT3-TKD (P = 0.02), MED12 (P = 0.02), and NPM1 (P = 0.02) mutations. They were younger (P = 0.007), had higher WBC (P = 0.001) and percentages of marrow (P < 0.001) and blood (P = 0.006) blasts, higher complete remission rates (P = 0.02), and longer overall survival (P < 0.001), thus indicating that atypical and typical CK-AMLs constitute distinct disease subtypes. We also identified smaller patient subsets within both typical and atypical CK-AML that differed molecularly and clinically.
Collapse
Affiliation(s)
- Krzysztof Mrózek
- 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 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 Comprehensive Cancer Center, Columbus, OH, USA
| | - James S Blachly
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Marius Bill
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | | | - Eunice S Wang
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Geoffrey L Uy
- Washington University School of Medicine in St. Louis, Siteman Cancer Center, St. Louis, MO, USA
| | - Jonathan E Kolitz
- Monter Cancer Center, Hofstra Northwell School of Medicine, Lake Success, NY, USA
| | - Bayard L Powell
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - William Blum
- Emory University School of Medicine, Atlanta, GA, USA
| | | | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Clara D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
6
|
Abstract
BACKGROUND Acute myelogenous leukemia (AML) is a hematopoietic neoplasm that primarily affects older adults. Despite scientific advances into the epidemiologic, genetic, and biological features of AML, this disease remains fatal to the majority of patients, particularly older individuals. METHODS We review the biologic and clinical characteristics of AML in the elderly and the treatment options that have emerged for them during the past several years. RESULTS Several biologic features of AML differ between older and younger individuals. Older patients often have disease that expresses multidrug resistance phenotype and cytogenetic abnormalities, which may be responsible in large part for the poor outcomes observed in older-aged subgroups. Traditional cytotoxic chemotherapy is associated with a low complete response rate and a high treatment-related mortality in older patients, which explains in part the poorer outcomes in cohorts over 60 years of age. Research into the pathophysiology of AML has revealed an abundance of intracellular signaling events that govern proliferation and survival of the malignant cell. Such discoveries have promoted recognition of new molecular and antigenic targets (eg, Flt-3 kinase, Ras, CD33 antigen) to which therapeutic development may be aimed. CONCLUSIONS New therapies directed against these unique targets may add to the current arsenal of antileukemic regimens and improve response rates and survival in older patients.
Collapse
|
7
|
Prognostic and biological significance of the proangiogenic factor EGFL7 in acute myeloid leukemia. Proc Natl Acad Sci U S A 2017; 114:E4641-E4647. [PMID: 28533390 DOI: 10.1073/pnas.1703142114] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epithelial growth factor-like 7 (EGFL7) is a protein that is secreted by endothelial cells and plays an important role in angiogenesis. Although EGFL7 is aberrantly overexpressed in solid tumors, its role in leukemia has not been evaluated. Here, we report that levels of both EGFL7 mRNA and EGFL7 protein are increased in blasts of patients with acute myeloid leukemia (AML) compared with normal bone marrow cells. High EGFL7 mRNA expression associates with lower complete remission rates, and shorter event-free and overall survival in older (age ≥60 y) and younger (age <60 y) patients with cytogenetically normal AML. We further show that AML blasts secrete EGFL7 protein and that higher levels of EGFL7 protein are found in the sera from AML patients than in sera from healthy controls. Treatment of patient AML blasts with recombinant EGFL7 in vitro leads to increases in leukemic blast cell growth and levels of phosphorylated AKT. EGFL7 blockade with an anti-EGFL7 antibody reduced the growth potential and viability of AML cells. Our findings demonstrate that increased EGFL7 expression and secretion is an autocrine mechanism supporting growth of leukemic blasts in patients with AML.
Collapse
|
8
|
The mutational oncoprint of recurrent cytogenetic abnormalities in adult patients with de novo acute myeloid leukemia. Leukemia 2017; 31:2211-2218. [PMID: 28321123 PMCID: PMC5628133 DOI: 10.1038/leu.2017.86] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 02/16/2017] [Indexed: 12/18/2022]
Abstract
Recurrent chromosomal abnormalities and gene mutations detected at the time of diagnosis of acute myeloid leukemia (AML) are associated with particular disease features, treatment response and survival of AML patients, and are used to denote specific disease entities in the World Health Organization classification of myeloid neoplasms and acute leukemia. However, large studies that integrate cytogenetic and comprehensive mutational information are scarce. We created a comprehensive oncoprint of mutations associated with recurrent cytogenetic findings by combining the information on mutational patterns of 80 cancer- and leukemia-associated genes with cytogenetic findings in 1603 adult patients with de novo AML. We show unique differences in the mutational profiles among major cytogenetic subsets, identify novel associations between recurrent cytogenetic abnormalities and both specific gene mutations and gene functional groups, and reveal differences in cytogenetic and mutational features between patients younger than 60 years and those aged 60 years or older. The identified associations between cytogenetic and molecular genetic data may help guide mutation testing in AML, and result in more focused application of targeted therapy in patients with de novo AML.
Collapse
|
9
|
Discovery of 1-hydroxypyridine-2-thiones as selective histone deacetylase inhibitors and their potential application for treating leukemia. Bioorg Med Chem Lett 2015; 25:4320-4. [DOI: 10.1016/j.bmcl.2015.07.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 07/16/2015] [Accepted: 07/21/2015] [Indexed: 11/19/2022]
|
10
|
Niederwieser C, Kohlschmidt J, Volinia S, Whitman SP, Metzeler KH, Eisfeld AK, Maharry K, Yan P, Frankhouser D, Becker H, Schwind S, Carroll AJ, Nicolet D, Mendler JH, Curfman JP, Wu YZ, Baer MR, Powell BL, Kolitz JE, Moore JO, Carter TH, Bundschuh R, Larson RA, Stone RM, Mrózek K, Marcucci G, Bloomfield CD. Prognostic and biologic significance of DNMT3B expression in older patients with cytogenetically normal primary acute myeloid leukemia. Leukemia 2015; 29:567-75. [PMID: 25204569 PMCID: PMC4351165 DOI: 10.1038/leu.2014.267] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/26/2014] [Accepted: 08/29/2014] [Indexed: 02/06/2023]
Abstract
DNMT3B encodes a DNA methyltransferase implicated in aberrant epigenetic changes contributing to leukemogenesis. We tested whether DNMT3B expression, measured by NanoString nCounter assay, associates with outcome, gene and microRNA expression and DNA methylation profiles in 210 older (⩾60 years) adults with primary, cytogenetically normal acute myeloid leukemia (CN-AML). Patients were dichotomized into high versus low expressers using median cut. Outcomes were assessed in the context of known CN-AML prognosticators. Gene and microRNA expression, and DNA methylation profiles were analyzed using microarrays and MethylCap-sequencing, respectively. High DNMT3B expressers had fewer complete remissions (CR; P=0.002) and shorter disease-free (DFS; P=0.02) and overall (OS; P<0.001) survival. In multivariable analyses, high DNMT3B expression remained an independent predictor of lower CR rates (P=0.04) and shorter DFS (P=0.04) and OS (P=0.001). High DNMT3B expression associated with a gene expression profile comprising 363 genes involved in differentiation, proliferation and survival pathways, but with only four differentially expressed microRNAs (miR-133b, miR-148a, miR-122, miR-409-3p) and no differential DNA methylation regions. We conclude that high DNMT3B expression independently associates with adverse outcome in older CN-AML patients. Gene expression analyses suggest that DNMT3B is involved in the modulation of several genes, although the regulatory mechanisms remain to be investigated to devise therapeutic approaches specific for these patients.
Collapse
MESH Headings
- Age Factors
- Aged
- Aged, 80 and over
- Cytarabine/therapeutic use
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA Methylation
- Daunorubicin/therapeutic use
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Humans
- Induction Chemotherapy
- Karyotyping
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Male
- MicroRNAs/genetics
- Microarray Analysis
- Middle Aged
- Prognosis
- Survival Analysis
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Alliance for Clinical Trials in Oncology Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Stefano Volinia
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Susan P. Whitman
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | | | - Kati Maharry
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Alliance for Clinical Trials in Oncology Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Pearlly Yan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | - Heiko Becker
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | | | | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
- Alliance for Clinical Trials in Oncology Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Jason H. Mendler
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - John P. Curfman
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Yue-Zhong Wu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Maria R. Baer
- Greenebaum Cancer Center, University of Maryland, Baltimore, MD
| | - Bayard L. Powell
- Comprehensive Cancer Center of Wake Forest University, Winston-Salem, NC
| | - Jonathan E. Kolitz
- Monter Cancer Center, Hofstra North Shore-Long Island Jewish School of Medicine, Lake Success, NY
| | | | | | - Ralf Bundschuh
- Departments of Physics and Chemistry & Biochemistry, The Ohio State University, Columbus, OH
| | | | | | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Guido Marcucci
- The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | | |
Collapse
|
11
|
Pastore F, Dufour A, Benthaus T, Metzeler KH, Maharry KS, Schneider S, Ksienzyk B, Mellert G, Zellmeier E, Kakadia PM, Unterhalt M, Feuring-Buske M, Buske C, Braess J, Sauerland MC, Heinecke A, Krug U, Berdel WE, Buechner T, Woermann B, Hiddemann W, Bohlander SK, Marcucci G, Spiekermann K, Bloomfield CD, Hoster E. Combined molecular and clinical prognostic index for relapse and survival in cytogenetically normal acute myeloid leukemia. J Clin Oncol 2014; 32:1586-94. [PMID: 24711548 DOI: 10.1200/jco.2013.52.3480] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Cytogenetically normal (CN) acute myeloid leukemia (AML) is the largest and most heterogeneous cytogenetic AML subgroup. For the practicing clinician, it is difficult to summarize the prognostic information of the growing number of clinical and molecular markers. Our purpose was to develop a widely applicable prognostic model by combining well-established pretreatment patient and disease characteristics. PATIENTS AND METHODS Two prognostic indices for CN-AML (PINA), one regarding overall survival (OS; PINAOS) and the other regarding relapse-free survival (RFS; PINARFS), were derived from data of 572 patients with CN-AML treated within the AML Cooperative Group 99 study (www.aml-score.org). RESULTS On the basis of age (median, 60 years; range, 17 to 85 years), performance status, WBC count, and mutation status of NPM1, CEBPA, and FLT3-internal tandem duplication, patients were classified into the following three risk groups according to PINAOS and PINARFS: 29% of all patients and 32% of 381 responding patients had low-risk disease (5-year OS, 74%; 5-year RFS, 55%); 56% of all patients and 39% of responding patients had intermediate-risk disease (5-year OS, 28%; 5-year RFS, 27%), and 15% of all patients and 29% of responding patients had high-risk disease (5-year OS, 3%; 5-year RFS, 5%), respectively. PINAOS and PINARFS stratified outcome within European LeukemiaNet genetic groups. Both indices were confirmed on independent data from Cancer and Leukemia Group B/Alliance trials. CONCLUSION We have developed and validated, to our knowledge, the first prognostic indices specifically designed for adult patients of all ages with CN-AML that combine well-established molecular and clinical variables and that are easily applicable in routine clinical care. The integration of both clinical and molecular markers could provide a basis for individualized patient care through risk-adapted therapy of CN-AML.
Collapse
Affiliation(s)
- Friederike Pastore
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand.
| | - Annika Dufour
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Tobias Benthaus
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Klaus H Metzeler
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Kati S Maharry
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Stephanie Schneider
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Bianka Ksienzyk
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Gudrun Mellert
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Evelyn Zellmeier
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Purvi M Kakadia
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Michael Unterhalt
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Michaela Feuring-Buske
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Christian Buske
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Jan Braess
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Maria Cristina Sauerland
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Achim Heinecke
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Utz Krug
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Wolfgang E Berdel
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Thomas Buechner
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Bernhard Woermann
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Wolfgang Hiddemann
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Stefan K Bohlander
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Guido Marcucci
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Karsten Spiekermann
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Clara D Bloomfield
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| | - Eva Hoster
- Friederike Pastore, Annika Dufour, Tobias Benthaus, Klaus H. Metzeler, Stephanie Schneider, Bianka Ksienzyk, Gudrun Mellert, Evelyn Zellmeier, Purvi M. Kakadia, Michael Unterhalt, Wolfgang Hiddemann, Stefan K. Bohlander, Karsten Spiekermann, and Eva Hoster, University Hospital Munich Großhadern; Friederike Pastore, Klaus H. Metzeler, Wolfgang Hiddemann, Stefan K. Bohlander, and Karsten Spiekermann, Helmholtz Center Munich; Eva Hoster, University of Munich, Munich; Purvi M. Kakadia and Stefan K. Bohlander, University Hospital Marburg, Marburg; Michaela Feuring-Buske, University Hospital Ulm; Christian Buske, Comprehensive Cancer Center Ulm, University of Ulm, Ulm; Jan Braess, Klinikum Barmherzige Brüder, Regensburg; Maria Cristina Sauerland and Achim Heinecke, University of Muenster; Utz Krug, Wolfgang E. Berdel, and Thomas Buechner, University Hospital Muenster, Muenster; Bernhard Woermann, German Society of Hematology and Oncology, Berlin, Germany; Kati S. Maharry, Guido Marcucci, and Clara D. Bloomfield, The Ohio State University Comprehensive Cancer Center, Columbus, OH; Kati S. Maharry, Mayo Clinic, Rochester, MN; and Stefan K. Bohlander, University of Auckland, Auckland, New Zealand
| |
Collapse
|
12
|
Massey PR, Fojo T, Bates SE. ABC Transporters: Involvement in Multidrug Resistance and Drug Disposition. CANCER DRUG DISCOVERY AND DEVELOPMENT 2014. [DOI: 10.1007/978-1-4614-9135-4_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
13
|
GAS6 expression identifies high-risk adult AML patients: potential implications for therapy. Leukemia 2013; 28:1252-1258. [PMID: 24326683 PMCID: PMC4047202 DOI: 10.1038/leu.2013.371] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/04/2013] [Accepted: 12/06/2013] [Indexed: 12/21/2022]
Abstract
Emerging data demonstrate important roles for the TYRO3/AXL/MERTK receptor tyrosine kinase (TAM RTK) family in diverse cancers. We investigated the prognostic relevance of GAS6 expression, encoding the common TAM RTK ligand, in 270 adults (n=71 aged <60 years; n=199 aged ≥60 years) with de novo cytogenetically normal acute myeloid leukemia (CN-AML). Patients expressing GAS6 (GAS6+), especially those aged ≥60 years, more often failed to achieve a complete remission (CR). In all patients, GAS6+ patients had shorter disease-free (DFS) and overall (OS) survival than patients without GAS6 expression (GAS6−). After adjusting for other prognostic markers, GAS6+ predicted CR failure (P=0.02), shorter DFS (P=0.004) and OS (P=0.04). To gain further biologic insights, we derived a GAS6-associated gene-expression signature (P<0.001) that in GAS6+ patients included overexpressed BAALC and MN1, known to confer adverse prognosis in CN-AML, and overexpressed CXCL12, encoding stromal cell-derived factor, and its receptor genes, CXCR4 and CXCR7. This study reports for the first time that GAS6 expression is an adverse prognostic marker in CN-AML. Although GAS6 decoy receptors are not yet available in the clinic for GAS6+ CN-AML therapy, potential alternative therapies targeting GAS6+-associated pathways, e.g., CXCR4 antagonists may be considered for GAS6+ patients to sensitize them to chemotherapy.
Collapse
|
14
|
Wetzler M, Mrózek K, Kohlschmidt J, Dombret H, Döhner H, Pilorge S, Krug U, Carroll AJ, Larson RA, Marcucci G, Hiddemann W, Büchner T, Bloomfield CD. Intensive induction is effective in selected octogenarian acute myeloid leukemia patients: prognostic significance of karyotype and selected molecular markers used in the European LeukemiaNet classification. Haematologica 2013; 99:308-13. [PMID: 24097631 DOI: 10.3324/haematol.2013.092072] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We investigated whether octogenarian patients with acute myeloid leukemia enrolled onto Cooperative Group clinical trials and treated with intensive induction therapy could be cured, and whether karyotype and selected molecular markers had any prognostic significance in these patients. Among 138 patients with cytogenetic information, normal karyotype was the most common (47.1%) followed by complex karyotype (14.5%) and sole +8 (9.4%). Among these patients, the relapse-free survival rate at 1 year was 37% and 13% at 3 years, and the respective overall survival rates were 24% and 8%. Whereas the 90 patients who survived beyond 30 days had the same relapse-free survival rates, their 1-year and 3-year overall survival rates were 36% and 11%, respectively. Of the 66 patients surviving beyond 30 days who could be classified into European LeukemiaNet genetic groups, those in the intermediate-I group had better overall survival than patients in the adverse group (P=0.01). Among patients with cytogenetically normal acute myeloid leukemia who were tested for the European LeukemiaNet-associated molecular alterations, FLT3-internal tandem duplication and NPM1 mutations, it was found that FLT3-internal tandem duplication (detected in 29% of patients) did not associate with overall survival (P=0.31), whereas NPM1 mutations (30%) were associated with a significantly longer overall survival (P=0.002). We conclude that intensive induction is effective and indicated in selected octogenarians with acute myeloid leukemia, that their overall survival varies among the European LeukemiaNet genetic groups and that NPM1 mutations may be of prognostic significance among octogenarian patients with cytogenetically normal acute myeloid leukemia.
Collapse
|
15
|
Marcucci G, Maharry KS, Metzeler KH, Volinia S, Wu YZ, Mrózek K, Nicolet D, Kohlschmidt J, Whitman SP, Mendler JH, Schwind S, Becker H, Eisfeld AK, Carroll AJ, Powell BL, Kolitz JE, Garzon R, Caligiuri MA, Stone RM, Bloomfield CD. Clinical role of microRNAs in cytogenetically normal acute myeloid leukemia: miR-155 upregulation independently identifies high-risk patients. J Clin Oncol 2013; 31:2086-93. [PMID: 23650424 DOI: 10.1200/jco.2012.45.6228] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To evaluate the impact of miR-155 on the outcome of adults with cytogenetically normal (CN) acute myeloid leukemia (AML) in the context of other clinical and molecular prognosticators and to gain insight into the leukemogenic role of this microRNA. PATIENTS AND METHODS We evaluated 363 patients with primary CN-AML. miR-155 levels were measured in pretreatment marrow and blood by NanoString nCounter assays that quantified the expression of the encoding gene MIR155HG. All molecular prognosticators were assessed centrally. miR-155-associated gene and microRNA expression profiles were derived using microarrays. RESULTS Considering all patients, high miR-155 expression was associated with a lower complete remission (CR) rate (P < .001) and shorter disease-free survival (P = .001) and overall survival (OS; P < .001) after adjusting for age. In multivariable analyses, high miR-155 expression remained an independent predictor for a lower CR rate (P = .007) and shorter OS (P < .001). High miR-155 expressers had approximately 50% reduction in the odds of achieving CR and 60% increase in the risk of death compared with low miR-155 expressers. Although high miR-155 expression was not associated with a distinct microRNA expression profile, it was associated with a gene expression profile enriched for genes involved in cellular mechanisms deregulated in AML (eg, apoptosis, nuclear factor-κB activation, and inflammation), thereby supporting a pivotal and unique role of this microRNA in myeloid leukemogenesis. CONCLUSION miR-155 expression levels are associated with clinical outcome independently of other strong clinical and molecular predictors. The availability of emerging compounds with antagonistic activity to microRNAs in the clinic provides the opportunity for future therapeutic targeting of miR-155 in AML.
Collapse
Affiliation(s)
- Guido Marcucci
- The Ohio State University, Comprehensive Cancer Center, Biomedical Research Tower 460 W. 12th Ave, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mrózek K, Marcucci G, Nicolet D, Maharry KS, Becker H, Whitman SP, Metzeler KH, Schwind S, Wu YZ, Kohlschmidt J, Pettenati MJ, Heerema NA, Block AW, Patil SR, Baer MR, Kolitz JE, Moore JO, Carroll AJ, Stone RM, Larson RA, Bloomfield CD. Prognostic significance of the European LeukemiaNet standardized system for reporting cytogenetic and molecular alterations in adults with acute myeloid leukemia. J Clin Oncol 2012; 30:4515-23. [PMID: 22987078 DOI: 10.1200/jco.2012.43.4738] [Citation(s) in RCA: 316] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To evaluate the prognostic significance of the international European LeukemiaNet (ELN) guidelines for reporting genetic alterations in acute myeloid leukemia (AML). PATIENTS AND METHODS We analyzed 1,550 adults with primary AML, treated on Cancer and Leukemia Group B first-line trials, who had pretreatment cytogenetics and, for cytogenetically normal patients, mutational status of NPM1, CEBPA, and FLT3 available. We compared complete remission (CR) rates, disease-free survival (DFS), and overall survival (OS) among patients classified into the four ELN genetic groups (favorable, intermediate-I, intermediate-II, adverse) separately for 818 younger (age < 60 years) and 732 older (age ≥ 60 years) patients. RESULTS The percentages of younger versus older patients in the favorable (41% v 20%; P < .001), intermediate-II (19% v 30%; P < .001), and adverse (22% v 31%; P < .001) genetic groups differed. The favorable group had the best and the adverse group the worst CR rates, DFS, and OS in both age groups. Both intermediate groups had significantly worse outcomes than the favorable but better than the adverse group. Intermediate-I and intermediate-II groups in older patients had similar outcomes, whereas the intermediate-II group in younger patients had better OS but not better CR rates or DFS than the intermediate-I group. The prognostic significance of ELN classification was confirmed by multivariable analyses. For each ELN group, older patients had worse outcomes than younger patients. CONCLUSION The ELN classification clearly separates the genetic groups by outcome, supporting its use for risk stratification in clinical trials. Because they have different proportions of genetic alterations and outcomes, younger and older patients should be reported separately when using the ELN classification.
Collapse
Affiliation(s)
- Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210-1228, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Marcucci G, Metzeler KH, Schwind S, Becker H, Maharry K, Mrózek K, Radmacher MD, Kohlschmidt J, Nicolet D, Whitman SP, Wu YZ, Powell BL, Carter TH, Kolitz JE, Wetzler M, Carroll AJ, Baer MR, Moore JO, Caligiuri MA, Larson RA, Bloomfield CD. Age-related prognostic impact of different types of DNMT3A mutations in adults with primary cytogenetically normal acute myeloid leukemia. J Clin Oncol 2012; 30:742-50. [PMID: 22291079 DOI: 10.1200/jco.2011.39.2092] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To determine the frequency of DNMT3A mutations, their associations with clinical and molecular characteristics and outcome, and the associated gene- and microRNA-expression signatures in primary cytogenetically normal acute myeloid leukemia (CN-AML). PATIENTS AND METHODS Four hundred fifteen previously untreated adults were analyzed for DNMT3A mutations and established prognostic gene mutations and expression markers. Gene- and microRNA-expression profiles were derived using microarrays. RESULTS Younger (< 60 years; n = 181) and older (≥ 60 years; n = 234) patients had similar frequencies of DNMT3A mutations (35.3% v 33.3%). Missense mutations affecting arginine codon 882 (R882-DNMT3A) were more common (n = 92; 62%) than those affecting other codons (non-R882-DNMT3A). DNMT3A-mutated patients did not differ regarding complete remission rate, but had shorter disease-free survival (DFS; P = .03) and, by trend, overall survival (OS; P = .07) than DNMT3A-wild-type patients. In multivariable analyses, DNMT3A mutations remained associated with shorter DFS (P = .01), but not with shorter OS. When analyzed separately, the two DNMT3A mutation types had different significance by age group. Younger patients with non-R882-DNMT3A mutations had shorter DFS (P = .002) and OS (P = .02), whereas older patients with R882-DNMT3A mutations had shorter DFS (P = .005) and OS (P = .002) after adjustment for other clinical and molecular prognosticators. Gene- and microRNA-expression signatures did not accurately predict DNMT3A mutational status. CONCLUSION DNMT3A mutations are frequent in CN-AML, and their clinical significance seems to be age dependent. DNMT3A-R882 mutations are associated with adverse prognosis in older patients, and non-R882-DNMT3A mutations are associated with adverse prognosis in younger patients. Low accuracy of gene- and microRNA-expression signatures in predicting DNMT3A mutation status suggested that the role of these mutations in AML remains to be elucidated.
Collapse
Affiliation(s)
- Guido Marcucci
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Escalation of daunorubicin and addition of etoposide in the ADE regimen in acute myeloid leukemia patients aged 60 years and older: Cancer and Leukemia Group B Study 9720. Leukemia 2011; 25:800-7. [PMID: 21321569 DOI: 10.1038/leu.2011.9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Untreated de novo (n=421) and secondary (n=189) acute myeloid leukemia (AML) patients ≥60 years received intensified chemotherapy, including daunorubicin 60 mg/m(2) and etoposide 100 mg/m(2) during days 1, 2, 3 with cytarabine 100 mg/m(2) during days 1-7, with a second induction if needed and one consolidation course with these drugs and doses for 2, 2 and 5 days, respectively. In all, 287 (47%) achieved complete remission (CR), 136 (22%) died and 187 (31%) were non-responders. CR rates were 27, 44 and 52% for complex karyotypes, rare aberrations and neither (P<0.001), 52 and 37% for de novo and secondary AML (P=0.003), and 53 and 42% for age 60-69 and ≥70 years (P=0.015). In multivariable analysis, CR predictors included non-complex/non-rare karyotypes (P<0.001), de novo AML (P<0.001), better performance status (PS) (P<0.001) and younger age (P=0.001). Disease-free (DFS) and overall (OS) survival medians were 6.8 (95% CI: 6.2, 7.8) and 7.2 (95% CI: 6.4, 8.6) months. In multivariable analysis, DFS was shorter for complex karyotypes (P<0.001) and increasing white blood count (WBC) (P<0.001) and age (P=0.038), and OS for complex karyotypes (P<0.001), increasing WBC (P=0.001) and age (P<0.001), poorer PS (P<0.001) and secondary AML (P=0.010). Outcomes and prognostic factors were similar to those in previous Cancer and Leukemia Group B studies.
Collapse
|
19
|
Marcucci G, Maharry K, Wu YZ, Radmacher MD, Mrózek K, Margeson D, Holland KB, Whitman SP, Becker H, Schwind S, Metzeler KH, Powell BL, Carter TH, Kolitz JE, Wetzler M, Carroll AJ, Baer MR, Caligiuri MA, Larson RA, Bloomfield CD. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. J Clin Oncol 2010; 28:2348-55. [PMID: 20368543 PMCID: PMC2881719 DOI: 10.1200/jco.2009.27.3730] [Citation(s) in RCA: 592] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2009] [Accepted: 01/27/2010] [Indexed: 01/11/2023] Open
Abstract
PURPOSE To analyze the frequency and associations with prognostic markers and outcome of mutations in IDH genes encoding isocitrate dehydrogenases in adult de novo cytogenetically normal acute myeloid leukemia (CN-AML). PATIENTS AND METHODS Diagnostic bone marrow or blood samples from 358 patients were analyzed for IDH1 and IDH2 mutations by DNA polymerase chain reaction amplification/sequencing. FLT3, NPM1, CEBPA, WT1, and MLL mutational analyses and gene- and microRNA-expression profiling were performed centrally. Results IDH mutations were found in 33% of the patients. IDH1 mutations were detected in 49 patients (14%; 47 with R132). IDH2 mutations, previously unreported in AML, were detected in 69 patients (19%; 13 with R172 and 56 with R140). R172 IDH2 mutations were mutually exclusive with all other prognostic mutations analyzed. Younger age (< 60 years), molecular low-risk (NPM1-mutated/FLT3-internal tandem duplication-negative) IDH1-mutated patients had shorter disease-free survival than molecular low-risk IDH1/IDH2-wild-type (wt) patients (P = .046). R172 IDH2-mutated patients had lower complete remission rates than IDH1/IDH2wt patients (P = .007). Distinctive microarray gene- and microRNA-expression profiles accurately predicted R172 IDH2 mutations. The highest expressed gene and microRNAs in R172 IDH2-mutated patients compared with the IDH1/IDH2wt patients were APP (previously associated with complex karyotype AML) and miR-1 and miR-133 (involved in embryonal stem-cell differentiation), respectively. CONCLUSION IDH1 and IDH2 mutations are recurrent in CN-AML and have an unfavorable impact on outcome. The R172 IDH2 mutations, previously unreported in AML, characterize a novel subset of CN-AML patients lacking other prognostic mutations and associate with unique gene- and microRNA-expression profiles that may lead to the discovery of novel, therapeutically targetable leukemogenic mechanisms.
Collapse
Affiliation(s)
- Guido Marcucci
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Kati Maharry
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Yue-Zhong Wu
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Michael D. Radmacher
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Krzysztof Mrózek
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Dean Margeson
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Kelsi B. Holland
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Susan P. Whitman
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Heiko Becker
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Sebastian Schwind
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Klaus H. Metzeler
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Bayard L. Powell
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Thomas H. Carter
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Jonathan E. Kolitz
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Meir Wetzler
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Andrew J. Carroll
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Maria R. Baer
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Michael A. Caligiuri
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Richard A. Larson
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| | - Clara D. Bloomfield
- From the Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH; The Cancer and Leukemia Group B Statistical Center, Duke University Medical Center, Durham; Comprehensive Cancer Center, Wake Forest University, Winston-Salem, NC; University of Iowa, Iowa City, IA; North Shore University Hospital, Manhasset; Roswell Park Cancer Institute, Buffalo, NY; University of Alabama at Birmingham, Birmingham, AL; University of Maryland, Baltimore, MD; and University of Chicago, Chicago, IL
| |
Collapse
|
20
|
Mutations of the Wilms tumor 1 gene (WT1) in older patients with primary cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Blood 2010; 116:788-92. [PMID: 20442368 DOI: 10.1182/blood-2010-01-262543] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We previously reported the adverse prognostic impact of Wilms tumor 1 gene (WT1) mutations in younger adult cytogenetically normal acute myeloid leukemia (CN-AML). Here, we investigated 243 older (> or = 60 years) primary CN-AML patients. WT1 mutated (WT1mut) patients (7%) had FLT3-ITD more frequently (P < .001), lower hemoglobin (P = .01), higher white blood cell count (P = .03) and percentage blood blasts (P = .03), and a shorter overall survival (P = .08) than WT1 wild-type (WT1wt) patients. Comparing older and younger WT1mut patients, they had similar pretreatment characteristics and outcome. By contrast, among WT1wt CN-AML, younger patients had a significantly better outcome. A WT1 mutation-associated gene-expression signature, reported here for the first time, included CD96, a leukemia stem cell-specific marker, and genes involved in gene regulation (eg, MLL, PML, and SNRPN) and in proliferative and metabolic processes (eg, INSR, IRS2, and PRKAA1), supporting the role of mutated WT1 in deregulating multiple homeostatic processes. Our results indicate that WT1mut CN-AML represents a distinct entity with poor treatment response across age groups. This study has been registered at www.clinicaltrials.gov as #NCT00900224.
Collapse
|
21
|
Chauncey TR, Gundacker H, Shadman M, List AF, Dakhil SR, Erba HP, Slovak ML, Chen IM, Willman CL, Kopecky KJ, Appelbaum FR. Sequential phase II Southwest Oncology Group studies (S0112 and S0301) of daunorubicin and cytarabine by continuous infusion, without and with ciclosporin, in older patients with previously untreated acute myeloid leukaemia. Br J Haematol 2010; 148:48-58. [PMID: 19821823 PMCID: PMC2967366 DOI: 10.1111/j.1365-2141.2009.07919.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Attempts to overcome multi-drug resistance in acute myeloid leukaemia (AML) have been limited by toxicities. To investigate the effect of reducing peak drug levels, we performed sequential phase II studies using continuous infusion daunorubicin and cytarabine without (AD) and then with ciclosporin (ADC) in older patients with AML. Untreated patients (age 56+ years) received daunorubicin (45 mg/m2 per day for 3 d) and cytarabine (200 mg/m2 per day for 7 d), both by continuous infusion, without (S0112, 60 patients) and then with (S0301, 50 patients) the addition of ciclosporin. Complete response (CR) rates were 38% on S0112 and 44% on S0301. Fatal induction toxicities occurred in 17% and 12% respectively, arising primarily from infection and haemorrhage. Median overall and relapse-free survival was 7 and 8 months for AD respectively, and 6 and 14 months for ADC. Patients with phenotypic or functional P-glycoprotein had somewhat higher CR rates with ADC than AD, although confidence intervals overlapped. In these sequential trials, continuous infusion AD produced CR rates comparable to those with bolus daunorubicin. The addition of ciclosporin did not cause undue toxicities, produced a similar CR rate, and possibly improved relapse-free survival. Further correlate analyses did not identify a subpopulation specifically benefitting from the addition of ciclosporin.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cyclosporine/administration & dosage
- Cyclosporine/adverse effects
- Cytarabine/administration & dosage
- Cytarabine/adverse effects
- Daunorubicin/administration & dosage
- Daunorubicin/adverse effects
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Epidemiologic Methods
- Female
- Humans
- Infusions, Intravenous
- Leukemia, Myeloid, Acute/blood
- Leukemia, Myeloid, Acute/drug therapy
- Male
- Middle Aged
- Neoplasm Proteins/metabolism
- Treatment Outcome
Collapse
|
22
|
Burkhart CA, Watt F, Murray J, Pajic M, Prokvolit A, Xue C, Flemming C, Smith J, Purmal A, Isachenko N, Komarov PG, Gurova KV, Sartorelli AC, Marshall GM, Norris MD, Gudkov AV, Haber M. Small-molecule multidrug resistance-associated protein 1 inhibitor reversan increases the therapeutic index of chemotherapy in mouse models of neuroblastoma. Cancer Res 2009; 69:6573-80. [PMID: 19654298 DOI: 10.1158/0008-5472.can-09-1075] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The multidrug resistance-associated protein 1 (MRP1) has been closely linked to poor treatment response in several cancers, most notably neuroblastoma. Homozygous deletion of the MRP1 gene in primary murine neuroblastoma tumors resulted in increased sensitivity to MRP1 substrate drugs (vincristine, etoposide, and doxorubicin) compared with tumors containing both copies of wild-type MRP1, indicating that MRP1 plays a significant role in the drug resistance in this tumor type and defining this multidrug transporter as a target for pharmacologic suppression. A cell-based readout system was created to functionally determine intracellular accumulation of MRP1 substrates using a p53-responsive reporter as an indicator of drug-induced DNA damage. Screening of small-molecule libraries in this readout system revealed pyrazolopyrimidines as a prominent structural class of potent MRP1 inhibitors. Reversan, the lead compound of this class, increased the efficacy of both vincristine and etoposide in murine models of neuroblastoma (syngeneic and human xenografts). As opposed to the majority of inhibitors of multidrug transporters, Reversan was not toxic by itself nor did it increase the toxicity of chemotherapeutic drug exposure in mice. Therefore, Reversan represents a new class of nontoxic MRP1 inhibitor, which may be clinically useful for the treatment of neuroblastoma and other MRP1-overexpressing drug-refractory tumors by increasing their sensitivity to conventional chemotherapy.
Collapse
|
23
|
Pein F, Pinkerton R, Berthaud P, Pritchard-Jones K, Dick G, Vassal G. Dose finding study of oral PSC 833 combined with weekly intravenous etoposide in children with relapsed or refractory solid tumours. Eur J Cancer 2007; 43:2074-81. [PMID: 17716890 DOI: 10.1016/j.ejca.2007.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Revised: 06/25/2007] [Accepted: 07/04/2007] [Indexed: 10/22/2022]
Abstract
PSC 833 is an effective MDR1 reversal agent in vitro, including studies with paediatric cancer cell lines such as neuroblastoma and rhabdomyosarcoma. This study was performed to determine the safety profile, dose limiting toxicity (DLT) and maximum tolerated dose (MTD) in children with solid tumours and to determine the influence of PSC 833 on the pharmacokinetics of co-administered etoposide. Each patient received one cycle of intravenous etoposide (100 mg/m2 daily for 3 days on three consecutive weeks) to document baseline pharmacokinetics, and subsequently the same schedule using a dose of 50 mg/m2 was given combined with PSC 833 given orally every 6h at a starting dose of 4 mg/kg. Thirty two eligible patients (23 male, median age 8.3 years) were enrolled. Neuroblastoma and rhabdomyosarcoma were the common disease types. Brain tumours were excluded. DLT was defined as any non-haematological grade 3-4 toxicity (common toxicity criteria) and using a specific toxicity scale for cerebellar toxicity. The MDT was defined as the first dose below which 2 or more patients per dose level experienced DLT. Grade 1-2 ataxia occurred in cohorts 2 and 3 (4 and 5 mg/kg, respectively). Three patients developed grade 3 neurotoxicity in the 6 mg/kg cohort and this defined the MTD. Six responses were observed (2 CR, 4 PR). Pharmacokinetic studies indicated that the clearance of etoposide was reduced by approximately 50% when combined with PSC 833. It is concluded that the toxicity profile and MDT is similar in both children and adults, as is the effect on etoposide metabolism. The study demonstrated the feasibility and safety of carrying out a paediatric phase 1 trial across European boundaries and acts as a model for future cooperative studies in rare cancers among children.
Collapse
Affiliation(s)
- F Pein
- Institut Regional du Cancer Nantes Atlantique, Dept de Recherche Therapeutique, CLCC Rene Gauducheau, Nantes, France
| | | | | | | | | | | |
Collapse
|
24
|
Larson RA, Stone RM, Mayer RJ, Schiffer CA. Fifty Years of Clinical Research by the Leukemia Committee of the Cancer and Leukemia Group B. Clin Cancer Res 2006; 12:3556s-63s. [PMID: 16740785 DOI: 10.1158/1078-0432.ccr-06-9001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Progress in the care of patients with leukemia has been one of the great success stories in the field of oncology, and clinical research in leukemia has been the "flagship" of the Cancer and Leukemia Group B since the inception of this organization. Lessons learned from the founders' emphasis on childhood and adult leukemia have been extended broadly over the past 50 years to virtually all types of malignant diseases, and the Leukemia Committee has continued to provide leadership and key contributions. The Leukemia Committee is focused on the individualization of treatment based on distinctive biological and clinical characteristics with the aim of increasing efficacy and decreasing nonspecific toxicity. Our clinical trials in leukemia and myeloma have shifted from primarily empirically derived comparisons of different chemotherapeutic regimens to testing novel concepts such as the role of dose intensity, inhibition of specific mechanisms of drug resistance, the use of hematopoietic growth factors and monoclonal antibodies, and the utility of targeted agents. The Cancer and Leukemia Group B was the pioneer among the cooperative groups in the creation of centralized tissue repositories and the incorporation of correlative laboratory studies as an integral feature of clinical trials, a practice now termed "translational research." Considerable effort has focused on the identification of important pretreatment characteristics, such as morphologic features, immunophenotype, chromosomal abnormalities, and molecular defects, which are significantly associated with outcome in multivariable analyses and which enhance our understanding for the complex biology of these diseases.
Collapse
Affiliation(s)
- Richard A Larson
- Department of Medicine and Cancer Research Center, University of Chicago, Chicago, Illinois, USA.
| | | | | | | |
Collapse
|
25
|
Abstract
The experience of most collaborative study groups is that the outcome for older patients has, unlike in younger patients, failed to improve over the last two decades. In addition there are a substantial number of older patients who do not enter collaborative group trials because they are not considered suitable for an intensive chemotherapy approach. During this era many combinations of chemotherapeutic agents at different dose levels have been tried. It is clear that novel agents and new approaches must be used to improve the situation, and should include options for patients who are not fit for intensive treatment. Fortunately, the increased understanding of the molecular basis and heterogeneity of the disease has fostered the development of novel agents. Chemo-resistance is a key characteristic of acute myeloid leukaemia (AML) in older patients and a number of randomized trials have now been completed to assess this approach. New possibilities of selectively killing leukemic cells and/or modifying toxicity are in prospect with the development of antibody directed chemotherapy in the form of gemtuzumab ozogamicin (Mylotarg; Wyeth, Philadelphia, PA). New drugs such as clofarabine or cloretazine are being evaluated. Molecular mechanisms, whether recognized or not, have been targeted by the use of FLT-3 and farnesyl transferase (FT) inhibitors. With several new agents to evaluate, novel approaches to trial design aimed at detecting options likely to make a useful impact are needed.
Collapse
Affiliation(s)
- Alan K Burnett
- Department of Haematology, University Hospital of Wales, Cardiff, UK.
| | | |
Collapse
|
26
|
Farag SS, Archer KJ, Mrózek K, Ruppert AS, Carroll AJ, Vardiman JW, Pettenati MJ, Baer MR, Qumsiyeh MB, Koduru PR, Ning Y, Mayer RJ, Stone RM, Larson RA, Bloomfield CD. Pretreatment cytogenetics add to other prognostic factors predicting complete remission and long-term outcome in patients 60 years of age or older with acute myeloid leukemia: results from Cancer and Leukemia Group B 8461. Blood 2006; 108:63-73. [PMID: 16522815 PMCID: PMC1895823 DOI: 10.1182/blood-2005-11-4354] [Citation(s) in RCA: 232] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We investigated the relative prognostic significance of cytogenetics in 635 adult acute myeloid leukemia (AML) patients 60 years of age or older treated on front-line protocols. Classification trees and tree-structured survival analysis (TSSA) were used to identify important cytogenetic groups, and their prognostic significance was then assessed in multivariable analysis (MVA). Overall, 48.5% achieved complete remission (CR); 6.6% survived at 5 years. Complex karyotypes with at least 3 abnormalities (complex > or = 3) and a group including "rare aberrations" predicted lower CR rates (25% and 30%) versus other patients (56%). Compared with complex > or = 3, the odds of CR were significantly higher for noncomplex karyotypes without rare aberrations on MVA. Cytogenetically, complex > or = 5 predicted inferior disease-free survival on TSSA, remaining significant on MVA together with white blood cell count (WBC), sex, and age. For survival, complex > or = 5, rare aberrations, and core-binding factor (CBF) abnormalities were prognostic (P < .001), with 5-year survivals of 0%, 0%, and 19.4%, respectively, and 7.5% for remaining patients. Together with WBC, marrow blasts, sex, and age, the cytogenetic groups remained significant on MVA. In conclusion, pretreatment cytogenetics adds to other prognostic factors in older AML patients. Patients with complex > or = 5 appear to benefit minimally from current treatment and are better suited for investigational therapy or supportive care.
Collapse
|
27
|
Matsouka P, Pagoni M, Zikos P, Giannakoulas N, Apostolidis I, Asprogeraka T, Arvanitopoulou E, Spanoudakis E, Kotsianidis I, Tsatalas K, Papaioannou M, Marinakis T, Skandali A, Viniou N, Yataganas X, Bakiri M. Addition of cyclosporin-A to chemotherapy in secondary (post-MDS) AML in the elderly. A multicenter randomized trial of the Leukemia Working Group of the Hellenic Society of Hematology. Ann Hematol 2006; 85:250-6. [PMID: 16416114 DOI: 10.1007/s00277-005-0066-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Accepted: 12/06/2005] [Indexed: 10/25/2022]
Abstract
In elderly patients with secondary leukemia, poor therapeutic response and low overall survival have been attributed mainly to age and to the primary resistance of leukemic cells to chemotherapy. Modulation of resistance has been attempted in different studies, but the results have been contradictory. We conducted an open, randomized multicenter clinical trial involving patients more than 60 years old with secondary leukemia preceded by a myelodysplastic syndrome. The induction chemotherapy regimen included idarubicin, cytarabine, and etoposide (group A); randomization involved simultaneous administration of cyclosporin-A per os (group B). Fifty-five patients were evaluated, 26 in group A and 29 in group B. Overall complete remission was achieved in 40% of the patients, 27% vs 52% in groups A and B, respectively (p=0.01). Leukemia-free survival was more favorable in patients who received cyclosporin-A, 12 vs 7 months for groups B and A, respectively (p=0.03). In a follow up period of 30 months, 7 out of 55 patients (13%) were alive, 4 of whom were in complete remission. Five out of the 7 alive patients were randomized in group B and had received cyclosporin-A. Treatment failure was higher in group A [19 of 26 patients (73%)] than in group B with CsA [14 of 29 patients (48%)] (p<0.0001). Treatment-related toxicity/mortality was 13%. Modulation of drug resistance by CsA in elderly people suffering from secondary acute leukemia may improve the outcome of chemotherapy without increasing drug toxicity and treatment-related mortality.
Collapse
Affiliation(s)
- P Matsouka
- Hematology Division, Internal Medicine Department, University Hospital Patras, Patras, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Oyekunle AA, Kröger N, Zabelina T, Ayuk F, Schieder H, Renges H, Fehse N, Waschke O, Fehse B, Kabisch H, Zander AR. Allogeneic stem-cell transplantation in patients with refractory acute leukemia: a long-term follow-up. Bone Marrow Transplant 2005; 37:45-50. [PMID: 16258531 DOI: 10.1038/sj.bmt.1705207] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We examined retrospectively 44 patients with refractory acute leukemia (acute myeloid leukemia (AML)/acute lymphoblastic leukemia=25/19) who underwent allogeneic transplantation at our center between 11/1990 and 04/2004. The median leukemic blasts was 25% and age 28 years (range, 3-56). Twenty-one patients had untreated relapse, 13 failed reinduction, eight in partial remission and two aplastic. Conditioning was myeloablative using cyclophosphamide, busulfan, total-body irradiation and etoposide (Bu/Cy/VP, n=22; TBI/Cy/VP, n=17; others, n=5) followed by marrow or peripheral blood transplant (n=23/21) from unrelated or related donors (n=28/16). All patients had graft-versus-host disease (GVHD) prophylaxis with cyclosporin and methotrexate. One patient experienced late graft failure. Severe acute-GVHD and chronic-GVHD appeared in eight and 14 patients, respectively. Thirteen patients (30%) remain alive after a median of 25.3 months (range, 2.4-134.1); with 31 deaths, mostly from relapse (n=15) and infections (n=12). Overall survival (OS) and progression-free survival (PFS) at 5 years was 28 and 26%, respectively. OS and PFS were significantly better with blasts < or =20% and time to transplant < or =1 year while transplant-related mortality was less with the use of TBI. We conclude that patients with refractory leukemia can benefit from allogeneic BMT, especially with < or =20% marrow blast.
Collapse
MESH Headings
- Adolescent
- Adult
- Blast Crisis/complications
- Blast Crisis/mortality
- Blast Crisis/pathology
- Blast Crisis/therapy
- Busulfan/administration & dosage
- Child
- Child, Preschool
- Cyclophosphamide/administration & dosage
- Disease-Free Survival
- Female
- Graft vs Host Disease/etiology
- Graft vs Host Disease/prevention & control
- Humans
- Leukemia, Myeloid, Acute/complications
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Myeloablative Agonists/administration & dosage
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Retrospective Studies
- Stem Cell Transplantation
- Transplantation Conditioning
- Transplantation, Homologous
- Whole-Body Irradiation/methods
Collapse
Affiliation(s)
- A A Oyekunle
- Department of Bone Marrow Transplantation, University-Hospital Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Xu WL, Shen HL, Ao ZF, Chen BA, Xia W, Gao F, Zhang YN. Combination of tetrandrine as a potential-reversing agent with daunorubicin, etoposide and cytarabine for the treatment of refractory and relapsed acute myelogenous leukemia. Leuk Res 2005; 30:407-13. [PMID: 16219352 DOI: 10.1016/j.leukres.2005.08.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Accepted: 08/08/2005] [Indexed: 11/19/2022]
Abstract
The potential mechanism of the chemotherapy resistance in acute myeloid leukemia (AML) is the multidrug resistance (MDR-1) gene product P-glycoprotein (P-gp), which is often overexpressed in myeloblasts from acute myeloid leukemia. In a multicenter clinical trial, 38 patients with poor risk forms of AML were treated with tetrandrine (TET), a potent inhibitor of the MDR-1 efflux pump, combined with daunorubicin (DNR), etoposide and cytarabine (TET-DEC). Overall, post-chemotherapy marrow hypoplasia was achieved in 36 patients. Sixteen patients (42%) achieved complete remission or restored chronic phase, 9 achieved partial remission (PR) and 13 failed therapy. Toxicities included infection, myelosuppression, stomatitis, mucositis, cerebellar toxicity and reversible cardiotoxicity. There was no significant difference in response for P-gp-positive and -negative patients. P-gp function was assessed in 26 patients by flow cytometric analysis, TET-contained plasma-augmented DNR accumulation relative to pretreatment plasma in K562/A02 cells by a median value of 88+/-101% (range, 11-501%). However, there was no difference in DNR uptake between responding and non-responding patients. Our data showed that TET-DEC was relatively well tolerated in these patients with poor risk AML, and had encouraging antileukemic effects.
Collapse
Affiliation(s)
- Wen-Lin Xu
- Department of Hematology, The Affiliated People's Hospital, Jiangsu University, 8 Dianli Road, Zhenjiang 212002, PR China.
| | | | | | | | | | | | | |
Collapse
|
30
|
Marcucci G, Mrózek K, Ruppert AS, Maharry K, Kolitz JE, Moore JO, Mayer RJ, Pettenati MJ, Powell BL, Edwards CG, Sterling LJ, Vardiman JW, Schiffer CA, Carroll AJ, Larson RA, Bloomfield CD. Prognostic Factors and Outcome of Core Binding Factor Acute Myeloid Leukemia Patients With t(8;21) Differ From Those of Patients With inv(16): A Cancer and Leukemia Group B Study. J Clin Oncol 2005; 23:5705-17. [PMID: 16110030 DOI: 10.1200/jco.2005.15.610] [Citation(s) in RCA: 283] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Because both t(8;21) and inv(16) disrupt core binding factor (CBF) in acute myeloid leukemia (AML) and confer relatively favorable prognoses, these cytogenetic groups are often treated similarly. Recent studies, however, have shown different gene profiling for the two groups, underscoring potential biologic differences. Therefore, we sought to determine whether these two cytogenetic groups should also be considered separate entities from a clinical standpoint. Patients and Methods We analyzed 144 consecutive adults with t(8;21) and 168 with inv(16) treated on Cancer and Leukemia Group B front-line studies. We compared pretreatment features, probability of achieving complete remission (CR), overall survival (OS) and cumulative incidence of relapse (CIR) between the two groups. Results With a median follow-up of 6.4 years, for CBF AML as a whole, the CR rate was 88%, 5-year OS was 50% and CIR was 53%. After adjusting for covariates, patients with t(8;21) had shorter OS (hazard ratio [HR] = 1.5; P = .045) and survival after first relapse (HR = 1.7; P = .009) than patients with inv(16). Unexpectedly, race was an important predictor for t(8;21) AML, in that nonwhites failed induction more often (odds ratio = 5.7; P = .006) and had shorter OS than whites when certain secondary cytogenetic abnormalities were present. In patients with t(8;21) younger than 60 years, type of induction also correlated with relapse risk. For inv(16) AML, secondary cytogenetic abnormalities (especially +22) and male sex predicted better outcome. Conclusion When the prognostic impact of race, secondary cytogenetic abnormalities, sex, and response to salvage treatment is considered, t(8;21) and inv(16) AMLs seem to be distinct clinical entities and should be stratified and reported separately.
Collapse
Affiliation(s)
- Guido Marcucci
- Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, A433B Starling-Loving Hall, 320 W 10th Ave, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bauer KS, Karp JE, Garimella TS, Wu S, Tan M, Ross DD. A phase I and pharmacologic study of idarubicin, cytarabine, etoposide, and the multidrug resistance protein (MDR1/Pgp) inhibitor PSC-833 in patients with refractory leukemia. Leuk Res 2005; 29:263-71. [PMID: 15661261 DOI: 10.1016/j.leukres.2004.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2004] [Accepted: 07/01/2004] [Indexed: 11/19/2022]
Abstract
This study was conducted to define the maximum tolerated dose (MTD), dose limiting toxicity (DLT), and pharmacokinetics of idarubicin when administered with and without the P-glycoprotein inhibitor PSC-833 in combination with cytarabine, and etoposide. Fifteen patients with relapsed and refractory acute leukemia were enrolled and received cytarabine as a 7-day continuous infusion, with etoposide and idarubicin administered for any three consecutive days during the cytarabine infusion. Two hours prior to the second dose of idarubicin, PSC-833 administration was initiated. The pharmacokinetics of idarubicin alone and with PSC-833 was assessed at three idarubicin dose levels (6, 8 and 10 mg/m(2)). The MTD of idarubicin in this combination was 8 mg/(m(2) day) with a DLT of oral mucositis. The complete remission rate (on an intent-to-treat basis) for this regimen was 33%, with a median duration of 6 months. The clearance of idarubicin was 140 +/- 200 and 181 +/- 94.3 l/h for idarubicin alone and with PSC-833, respectively. The volume of distribution of the central compartment was 423 +/- 443 and 337 +/- 394 l for idarubicin alone and in combination with PSC-833, respectively. This combination including PSC-833 was well tolerated. Although a pharmacokinetic interaction might have been expected, PSC-833 did not significantly alter the disposition of idarubicin.
Collapse
Affiliation(s)
- Kenneth S Bauer
- Greenebaum Cancer Center, University of Maryland School of Pharmacy, Allied Health Building Suite 540, 100 Penn Street, Baltimore, MD 21201, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Kolitz JE, George SL, Dodge RK, Hurd DD, Powell BL, Allen SL, Velez-Garcia E, Moore JO, Shea TC, Hoke E, Caligiuri MA, Vardiman JW, Bloomfield CD, Larson RA. Dose escalation studies of cytarabine, daunorubicin, and etoposide with and without multidrug resistance modulation with PSC-833 in untreated adults with acute myeloid leukemia younger than 60 years: final induction results of Cancer and Leukemia Group B Study 9621. J Clin Oncol 2004; 22:4290-301. [PMID: 15514371 DOI: 10.1200/jco.2004.11.106] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE P-glycoprotein (Pgp) is strongly inhibited by PSC-833. A chemotherapy dose-escalation study was performed with PSC-833 in patients younger than 60 years with untreated acute myeloid leukemia. Clinical rather than pharmacokinetic end points were used to develop two induction therapies containing drugs susceptible to Pgp-mediated efflux and associated with comparable toxicities at the maximum-tolerated doses. PATIENTS AND METHODS A total of 410 patients were enrolled. Fifteen induction regimens containing variable doses of daunorubicin (DNR) and etoposide (ETOP) and fixed doses of cytarabine were evaluated with (ADEP) or without (ADE) a fixed dose of PSC-833. RESULTS Doses selected for phase III testing were DNR 90 mg/m(2) and ETOP 100 mg/m(2) in ADE, and DNR and ETOP each 40 mg/m(2) in ADEP. Intolerable mucosal toxicity occurred at higher doses of ADEP. Although the design of this study precludes direct comparisons, there was an apparent advantage for receiving ADEP with respect to disease-free and overall survival in patients < or = 45 years old, despite the significantly lower doses of DNR and ETOP given in ADEP compared with ADE. CONCLUSION A large clinical data set was used to develop induction regimens containing two drugs susceptible to Pgp-mediated efflux, with and without an inhibitor of Pgp function. The chosen doses have comparable antileukemia activity and toxicity, making them suitable for use in a phase III comparative study of induction chemotherapy for patients with acute myeloid leukemia younger than 60 years. That trial will also clarify whether patients < or = 45 years old are especially likely to benefit from Pgp inhibition during induction therapy.
Collapse
Affiliation(s)
- Jonathan E Kolitz
- Don Monti Division of Oncology and Division of Hematology, Department of Medicine, North Shore University Hospital, New York University School of Medicine, NY 11030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Marcucci G, Mrózek K, Ruppert AS, Archer KJ, Pettenati MJ, Heerema NA, Carroll AJ, Koduru PRK, Kolitz JE, Sterling LJ, Edwards CG, Anastasi J, Larson RA, Bloomfield CD. Abnormal Cytogenetics at Date of Morphologic Complete Remission Predicts Short Overall and Disease-Free Survival, and Higher Relapse Rate in Adult Acute Myeloid Leukemia: Results From Cancer and Leukemia Group B Study 8461. J Clin Oncol 2004; 22:2410-8. [PMID: 15197203 DOI: 10.1200/jco.2004.03.023] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose As most patients with acute myeloid leukemia (AML) with morphologic complete remission (CR) ultimately relapse, better predictors for outcome are needed. Recently, Cheson et al suggested using cytogenetic remission (CRc) as part of the criteria for CR. To our knowledge, ours is the first relatively large study evaluating the usefulness of CRc attained immediately following induction chemotherapy. Patients and Methods We included AML patients treated on Cancer and Leukemia Group B front-line studies with cytogenetic samples obtained at diagnosis and at the first day of documented CR following induction. Patients with abnormal cytogenetics at diagnosis, and normal cytogenetics at CR (NCR; n = 103) were compared with those with abnormal cytogenetics both at diagnosis and at CR (ACR; n = 15) for overall survival (OS), disease-free survival (DFS), and cumulative incidence of relapse (CIR). Cox proportional hazards models determined the prognostic significance of cytogenetics at CR, adjusting for other covariates. Results Clinical features were similar for both groups, with the exception of favorable cytogenetics [t(8;21), inv(16)/t(16;16), t(15;17)] at diagnosis, which was more frequent (P = .03) in the NCR group. Median follow-up was 3.1 years (range, 1.0 to 11.4 years). ACR patients had significantly shorter OS (P = .006) and DFS (P = .0001), and higher CIR (P = .0001). In multivariable models, the NCR and ACR groups were predictors for OS (P = .03), DFS (P = .02), and CIR (P = .05). The relative risk of relapse or death was 2.1 times higher for ACR patients than for NCR patients (95% CI, 1.1 to 3.9). Conclusion Our data suggest that converting to normal karyotype at the time of first CR is an important prognostic indicator and support the use of CRc as a criterion of CR in AML.
Collapse
Affiliation(s)
- Guido Marcucci
- Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Drug resistance, intrinsic or acquired, is a problem for all chemotherapeutic agents. In this review, we examine numerous strategies that have been tested or proposed to reverse drug resistance. Included among these strategies are approaches targeting the apoptosis pathway. Although the process of apoptosis is complex, it provides several potential sites for therapeutic intervention. A variety of targets and approaches are being pursued, including the suppression of proteins inhibiting apoptosis using antisense oligonucleotides (ASOs), and small molecules targeted at proteins that modulate apoptosis. An alternate strategy is based on numerous studies that have documented methylation of critical regions in the genome in human cancers. Consequently, efforts have been directed at re-expressing genes, including genes that affect drug sensitivity, using 5-azacytidine and 2'-deoxy-5-azacytidine (DAC, decitabine) as demethylating agents. While this strategy may be effective as a single modality, success will most likely be achieved if it is used to modulate gene expression in combination with other modalities such as chemotherapy. At a more basic level, attempts have been made to modulate glutathione (GSH) levels. Owing to its reactivity and high intracellular concentrations, GSH has been implicated in resistance to several chemotherapeutic agents. Several approaches designed to deplete intracellular GSH levels have been pursued including the use of buthionine-(S,R)-sulfoxime (BSO), a potent and specific inhibitor of gamma-glutamyl cysteine synthetase (gamma-GCS), the rate-limiting step in the synthesis of GSH, a hammerhead ribozyme against gamma-GCS mRNA to downregulate specifically its levels and targeting cJun expression to reduce GSH levels. Alternate strategies have targeted p53. The frequent occurrence of p53 mutations in human cancer has led to the development of numerous approaches to restore wild-type (wt) p53. The goals of these interventions are to either revert the malignant phenotype or enhance drug sensitivity. The approach most extensively investigated has utilized one of several viral vectors. An alternate approach, the use of small molecules to restore wt function to mutant p53, remains an option. Finally, the conceptually simplest mechanism of resistance is one that reduces intracellular drug accumulation. Such reduction can be effected by a variety of drug efflux pumps, of which the most widely studied is P-glycoprotein (Pgp). The first strategy utilized to inhibit Pgp function relied on the identification of non-chemotherapeutic agents as competitors. Other approaches have included the use of hammerhead ribozymes against the MDR-1 gene and MDR-1-targeted ASOs. Although modulation of drug resistance has not yet been proven to be an effective clinical tool, we have learned an enormous amount about drug resistance. Should we succeed, these pioneering basic and clinical studies will have paved the road for future developments.
Collapse
Affiliation(s)
- Tito Fojo
- Center for Cancer Research, National Cancer Institute, Building 10, Room 12-C-103, 9000 Rockville Pike, Bethesda, MA 20892, USA.
| | | |
Collapse
|
35
|
Larson RA, Daley GQ, Schiffer CA, Porcu P, Pui CH, Marie JP, Steelman LS, Bertrand FE, McCubrey JA. Treatment by design in leukemia, a meeting report, Philadelphia, Pennsylvania, December 2002. Leukemia 2003; 17:2358-82. [PMID: 14562120 DOI: 10.1038/sj.leu.2403156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Novel approaches have been designed to treat leukemia based on our understanding of the genetic and biochemical lesions present in different malignancies. This meeting report summarizes some of the recent advances in leukemia treatment. Based on the discoveries of cellular oncogenes, chromosomal translocations, monoclonal antibodies, multidrug resistance pumps, signal transduction pathways, genomics/proteonomic approaches to clinical diagnosis and mutations in biochemical pathways, clinicians and basic scientists have been able to identify the particular genetic mutations and signal transduction pathways involved as well as design more appropriate treatments for the leukemia patient. This meeting report discusses these exciting new therapies and the results obtained from ongoing clinical trials. Furthermore, rational approaches to treat complications of tumor lysis syndrome by administration of the recombinant urate oxidase protein, also known as rasburicase, which corrects the biochemical defect present in humans, were discussed. Clearly, over the past 25 years, molecular biology and biotechnology has provided the hematologist/oncologist novel bullets in their arsenal that will allow treatment by design in leukemia.
Collapse
Affiliation(s)
- R A Larson
- Section of Hematology/Oncology, University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- R A Larson
- Department of Medicine and Cancer Research Center, Section of Hematology/Oncology, The University of Chicago, IL 60637, USA
| |
Collapse
|
37
|
Abstract
Acute myeloid leukaemia (AML) is a disease of the elderly (median age at presentation 64 years). The outcome in older patients with AML is much worse than that for similarly treated younger patients. Older patients have a high incidence of recognised poor prognostic features (poor performance status, unfavourable cytogenetics, CD34 positive phenotype, raised serum lactate dehydrogenase levels and increased incidence of multidrug resistance protein expression). In addition, treatment is less well tolerated as there is an increased incidence of comorbidity in the elderly. The outlook for most patients is poor (4% survival at 5 years). However, it is possible to select a group of patients who are fit, with no pre-existing problems and good performance status who will respond well to intensive chemotherapy, and these patients should be treated aggressively. Less intensive treatment is probably more suitable for patients not fitting these criteria. Patients and their relatives should be counselled appropriately as to the prognosis of AML, the choices of treatment available and that intensive regimens are not an appropriate choice for many patients.
Collapse
Affiliation(s)
- Graham H Jackson
- Department of Haematology, Royal Victoria Infirmary, Newcastle Upon Tyne, UK.
| | | |
Collapse
|
38
|
Phase 3 study of the multidrug resistance modulator PSC-833 in previously untreated patients 60 years of age and older with acute myeloid leukemia: Cancer and Leukemia Group B Study 9720. Blood 2002. [DOI: 10.1182/blood.v100.4.1224.h81602001224_1224_1232] [Citation(s) in RCA: 265] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Cancer and Leukemia Group B conducted a phase 3 trial of the P-glycoprotein modulator PSC-833 in untreated acute myeloid leukemia patients aged 60 years and older. Patients were randomized to 1 of 2 regimens, with doses determined in a prior phase 1 study, consisting of cytarabine 100 mg/m2/d by 7-day infusion, with daunorubicin 60 mg/m2 and etoposide 100 mg/m2 daily for 3 days (ADE), or daunorubicin 40 mg/m2 and etoposide 60 mg/m2 for 3 days with PSC-833, 2.8 mg/kg over 2 hours, and then 10 mg/kg/d by 3-day infusion (ADEP). The ADEP arm was closed after randomization of 120 patients (61 to ADE and 59 to ADEP) because of excessive early mortality. Rates of complete remission, nonresponse, and death were 46%, 34%, and 20% for ADE, versus 39%, 17%, and 44% for ADEP (P = .008). Nevertheless, disease-free survival (median 7 vs 8 months; P = .38) and overall survival (approximately 33% alive at 1 year) did not differ and were similar to historical results. Although the number of patients was limited, ADE patients whose pretreatment cells exhibited PSC-833–modulated dye efflux in vitro (n = 22) had worse outcomes than those without efflux (n = 11) (complete remission, nonresponse, and death rates of 41%, 41%, and 18%, compared with 91%, 9%, and 0%; P = .03), but with ADEP outcomes were nearly identical. Moreover, for patients with PSC-833–modulated efflux, median disease-free survival was 5 months with ADE and 14 months with ADEP (P = .07). Further modulation trials in older patients must await the design of less-toxic regimens.
Collapse
|
39
|
Stone RM. New therapies in leukemia: renewed hope. JOURNAL OF HEMATOTHERAPY & STEM CELL RESEARCH 2002; 11:583-7. [PMID: 12201947 DOI: 10.1089/15258160260194730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Richard M Stone
- Adult Leukemia Program, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA. rstone @partners.org
| |
Collapse
|
40
|
Successful Treatment of Angioinvasive Aspergillosis During Prolonged Neutropenia with Liposomal Amphotericin, Voriconazole, and Caspofungin. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2002. [DOI: 10.1097/01.idc.0000078754.71576.ce] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Schaich M, Illmer T. Mdr1 gene expression and mutations in Ras proto-oncogenes in acute myeloid leukemia. Leuk Lymphoma 2002; 43:1345-54. [PMID: 12389613 DOI: 10.1080/10428190290033279] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Resistance to cytotoxic therapy and development of refractory disease in acute myeloid leukemia (AML) is frequently associated with the expression of mdr1/P-gp. In the last years many potential signaling pathways leading to modulation of mdr1 expression have been described. Thus, it has been assumed that activated Ras may influence mdr1 expression. This activation can be realized by mutations in the Ras oncogene leading to constitutive signaling. Ras mutations are observed in many human cancers, including AML. Recently, we could show a negative correlation between Ras mutations and mdr1 expression in blast samples of AML patients. Taking this up the potential possibilities of Ras influence on mdr1 activity and their implications on treatment outcome in AML are discussed.
Collapse
Affiliation(s)
- Markus Schaich
- Department of Medicine I, University Hospital C.G. Carus, Dresden, Germany.
| | | |
Collapse
|
42
|
van der Kolk DM, de Vries EGE, Müller M, Vellenga E. The role of drug efflux pumps in acute myeloid leukemia. Leuk Lymphoma 2002; 43:685-701. [PMID: 12153153 DOI: 10.1080/10428190290016773] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
A major problem in the treatment of patients with acute myeloid leukemia (AML) is the occurrence of resistance to structurally and functionally unrelated chemotherapeutic agents, called multidrug resistance (MDR). One of the known MDR mechanisms is the overexpression of adenosine triphosphate (ATP)-dependent efflux pumps. Permeability-glycoprotein (P-gp), the best characterized of the human drug efflux pumps, has been shown to be associated with poor treatment outcome in AML patients. Besides P-gp, in addition the multidrug resistance protein 1 (MRP1) appeared to contribute to the observed resistance in AML. Alternative transporter proteins, such as the MRP1 homologues MRP2, MRP3, MRP5 and MRP6, and the breast cancer resistance protein (BCRP), have been shown to be expressed at variable levels in AML patient cells. The latter proteins have been described to confer resistance to chemotherapeutic agents, such as daunorubicin, mitoxantrone, etoposide and 6-mercaptopurine, which are generally used in the treatment of AML patients; however, theyhave not yet proven to play a role in drug resistance in AML. The present review gives an overview of the current knowledge concerning these drug transporters, with a focus on the role of the transporter proteins in AML.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/analysis
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/physiology
- Animals
- Biological Transport
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Hematopoiesis
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Mice
- Neoplasm Proteins
Collapse
|
43
|
Schiffer CA. Postremission therapy in older adults with acute myeloid leukemia: an opportunity for new drug development. Leukemia 2002; 16:745-7. [PMID: 11960360 DOI: 10.1038/sj.leu.2402487] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2001] [Accepted: 01/24/2002] [Indexed: 11/09/2022]
Affiliation(s)
- C A Schiffer
- Karmanos Cancer Institute and the Division of Hematology/Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
44
|
Ma MK, McLeod HL, Westervelt P, Fracasso PM. Pharmacokinetic Study of Infusional Valspodar. J Clin Pharmacol 2002. [DOI: 10.1177/00912700222011463] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Burnett AK. Acute myeloid leukemia: treatment of adults under 60 years. REVIEWS IN CLINICAL AND EXPERIMENTAL HEMATOLOGY 2002; 6:26-45; discussion 86-7. [PMID: 12060482 DOI: 10.1046/j.1468-0734.2002.00058.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Current chemotherapy with advanced supportive care will enable 75-80% of acute myeloid leukemia patients aged 60 years or under to enter complete remission. Several regimens achieve broadly similar results. For patients who enter complete remission, the overall relapse risk is now 45-50%, but this is highly variable and is primarily determined by the biology of the disease. Cytogenetics are strongly influential in response to induction and consolidation with t(15:17), t(8:21) and inv(16) either occurring alone or with additional abnormalities having a relapse risk of about 30% and complex changes, abnormal 3q or abnormalities of chromosomes 5 and 7 resulting in a lower remission rate and a rapid relapse cumulating to 80%. FLT3 mutations occur in 25% of patients and are an independent predictor of relapse and, when combined with cytogenetics, adversely influence the prognosis in each cytogenetic risk group. Recent prospective collaborative group trials have endeavored to evaluate allogeneic and autologous bone marrow transplant against or in addition to consolidation chemotherapy. Suboptimal treatment delivery emerged as a problem. When the results were reported on an intention-to-treat basis, no overall survival advantage was consistently seen for either type of transplant. However, a significant reduction in risk of relapse was usually seen overall and within risk groups. Analysis within risk groups suggests that transplant is not indicated in good risk disease and continues to require evaluation in standard or poor risk patients. It is probable that traditional dose intensification has now reached its limits of tolerability, so new approaches will be required for further progress to be made. Modulation of chemoresistance mechanisms or immunologically directed chemotherapy represent immediate prospects for clinical study.
Collapse
Affiliation(s)
- Alan K Burnett
- Department of Hematology, University of Wales College of Medicine, Heath Park, Cardiff, UK.
| |
Collapse
|
46
|
Abstract
Chemotherapeutics are the most effective treatment for metastatic tumours. However, the ability of cancer cells to become simultaneously resistant to different drugs--a trait known as multidrug resistance--remains a significant impediment to successful chemotherapy. Three decades of multidrug-resistance research have identified a myriad of ways in which cancer cells can elude chemotherapy, and it has become apparent that resistance exists against every effective drug, even our newest agents. Therefore, the ability to predict and circumvent drug resistance is likely to improve chemotherapy.
Collapse
Affiliation(s)
- Michael M Gottesman
- Laboratory of Cell Biology and Cancer Therapeutics Branch, The Center for Cancer Research, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | |
Collapse
|
47
|
|
48
|
Uchiyama-Kokubu N, Watanabe T, Cohen D. Intracellular levels of two cyclosporin derivatives valspodar (PSC 833) and cyclosporin a closely associated with multidrug resistance-modulating activity in sublines of human colorectal adenocarcinoma HCT-15. Jpn J Cancer Res 2001; 92:1116-26. [PMID: 11676863 PMCID: PMC5926616 DOI: 10.1111/j.1349-7006.2001.tb01067.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
P-Glycoprotein, which mediates multidrug resistance (MDR) in cancer chemotherapy, is a principal target of cyclosporin A and [3'-keto-Bmt(1)]-[Val(2)]-cyclosporin (valspodar; PSC 833). To clarify mechanisms contributing to the different MDR-modulating activities of valspodar and cyclosporin A, we investigated the relation of the intracellular levels of the two cyclosporin derivatives to their modulating effect on MDR in different P-glycoprotein-expressing human colorectal carcinoma HCT-15 cells (parental HCT-15 and adriamycin-resistant sublines). In this study, valspodar was found to be much more potent than cyclosporin A in both sensitizing resistant cells to MDR-related anticancer drugs (e.g., adriamycin, vincristine and paclitaxel (taxol)) and increasing 2-[6-amino-3-imino-3H-xanthen-9-yl]benzoic acid methyl ester (rhodamine 123) retention and [G-(3)H]vincristine sulfate ([(3)H]vincristine) accumulation in these cells. Furthermore, a good correlation was detected between P-glycoprotein levels and the MDR-reversing effect of valspodar. In contrast, the effects of cyclosporin A could not be linked to P-glycoprotein levels in the MDR cells. In addition, the intracellular accumulation of valspodar was found to be 3 - 6 fold higher than that of cyclosporin A in four sublines and verapamil, an inhibitor of P-glycoprotein-mediated transport, enhanced the accumulation of cyclosporin A, but not valspodar. These results suggested that valspodar accumulation is not actively regulated by the P-glycoprotein-mediated efflux system.
Collapse
Affiliation(s)
- N Uchiyama-Kokubu
- Tsukuba Research Institute, Novartis Pharma K.K., Ohkubo 8, Tsukuba, Ibaraki 300-2611.
| | | | | |
Collapse
|
49
|
Stone RM, Berg DT, George SL, Dodge RK, Paciucci PA, Schulman PP, Lee EJ, Moore JO, Powell BL, Baer MR, Bloomfield CD, Schiffer CA. Postremission therapy in older patients with de novo acute myeloid leukemia: a randomized trial comparing mitoxantrone and intermediate-dose cytarabine with standard-dose cytarabine. Blood 2001; 98:548-53. [PMID: 11468148 DOI: 10.1182/blood.v98.3.548] [Citation(s) in RCA: 162] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The treatment of older patients with acute myeloid leukemia (AML) remains unsatisfactory, with complete remission (CR) achieved in only approximately 50% and long-term disease-free survival in 10% to 20%. Three hundred eighty-eight patients (60 years of age and older) with newly diagnosed de novo AML were randomly assigned to receive placebo (P) or granulocyte-macrophage colony-stimulating factor (GM-CSF) or GM in a double-blind manner, beginning 1 day after the completion of 3 days of daunorubicin and 7 days of cytarabine therapy. No differences were found in the rates of leukemic regrowth, CR, or infectious complications in either arm. Of 205 patients who achieved CR, 169 were medically well and were randomized to receive cytarabine alone or a combination of cytarabine and mitoxantrone. With a median follow-up of 7.7 years, the median disease-free survival times were 11 months and 10 months for those randomized to cytarabine or cytarabine/mitoxantrone, respectively. Rates of relapse, excluding deaths in CR, were 77% for cytarabine and 82% for cytarabine/mitoxantrone. Induction randomization had no effect on leukemic relapse rate or remission duration in either postremission arm. Because cytarabine/mitoxantrone was more toxic and no more effective than cytarabine, it was concluded that this higher-dose therapy had no benefit in the postremission management of older patients with de novo AML. These results suggest the need to develop novel therapeutic strategies for these patients. (Blood. 2001;98:548-553)
Collapse
Affiliation(s)
- R M Stone
- Dana-Farber Cancer Institute, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
A patient with acute myeloid leukemia (AML) who has achieved remission after induction chemotherapy still harbors 10(9) to 10(10), albeit undetectable, leukemic cells. Optimally, postremission therapy safely reduces the leukemic burden to a level compatible with long-term disease-free survival. Although older adults fare poorly with intensive postremission therapy, young and middle-aged adults can receive either chemotherapy based on high-dose cytarabine or myeloablative therapy in preparation for autologous or, if a histocompatible donor is available, allogeneic stem cell rescue. The rationale for each approach, the prospective trials comparing the various options, and a suggested strategy for choosing among them is presented. Although cytogenetic category at diagnosis is the most important prognostic factor, this feature remains an imperfect guide to postremission therapy. An informal consensus has arisen in favor of chemotherapy for patients with good prognosis and allogeneic transplant for those whose AML displays an adverse karyotype. For the intermediate group, an individualized decision is required and any of the three options is reasonable. Because the relapse rate is so high, new therapies for AML, such as signaling and immunotherapeutic approaches, are the focus of active investigation.
Collapse
Affiliation(s)
- R M Stone
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| |
Collapse
|