229 results on '"Yasunobu Nagata"'
Search Results
2. Distinct clinical and biological implications of CUX1 in myeloid neoplasms
- Author
-
Mai Aly, Zubaidah M. Ramdzan, Yasunobu Nagata, Suresh K. Balasubramanian, Naoko Hosono, Hideki Makishima, Valeria Visconte, Teodora Kuzmanovic, Vera Adema, Aziz Nazha, Bartlomiej P. Przychodzen, Cassandra M. Kerr, Mikkael A. Sekeres, Mohamed E. Abazeed, Alain Nepveu, and Jaroslaw P. Maciejewski
- Subjects
Specialties of internal medicine ,RC581-951 - Abstract
Abstract: Somatic mutations of the CUT-like homeobox 1 (CUX1) gene (CUX1MT) can be found in myeloid neoplasms (MNs), in particular, in myelodysplastic syndromes (MDSs). The CUX1 locus is also deleted in 3 of 4 MN cases with −7/del(7q). A cohort of 1480 MN patients was used to characterize clinical features and clonal hierarchy associated with CUX1MT and CUX1 deletions (CUX1DEL) and to analyze their functional consequences in vitro. CUX1MT were present in 4% of chronic MNs. CUX1DEL were preferentially found in advanced cases (6%). Most MDS and acute myeloid leukemia (AML) patients with −7/del(7q) and up to 15% of MDS patients and 5% of AML patients diploid for the CUX1 locus exhibited downmodulated CUX1 expression. In 75% of mutant cases, CUX1MT were heterozygous, whereas microdeletions and homozygous and compound-heterozygous mutations were less common. CUXMT/DEL were associated with worse survival compared with CUX1WT. Within the clonal hierarchy, 1 of 3 CUX1MT served as founder events often followed by secondary BCOR and ASXL1 subclonal hits, whereas TET2 was the most common ancestral lesion, followed by subclonal CUX1MT. Comet assay of patients' bone marrow progenitor cells and leukemic cell lines performed in various experimental conditions revealed that frameshift mutations, hemizygous deletions, or experimental CUX1 knockdown decrease the repair of oxidized bases. These functional findings may explain why samples with either CUX1MT or low CUX1 expression coincided with significantly higher numbers of somatic hits by whole-exome sequencing. Our findings implicate the DNA repair dysfunction resulting from CUX1 lesions in the pathogenesis of MNs, in which they lead to a mutator phenotype.
- Published
- 2019
- Full Text
- View/download PDF
3. Genomics of therapy-related myeloid neoplasms
- Author
-
Teodora Kuzmanovic, Bhumika J. Patel, Srinivasa R. Sanikommu, Yasunobu Nagata, Hassan Awada, Cassandra M. Kerr, Bartlomiej P. Przychodzen, Babal K. Jha, Devendra Hiwase, Deepak Singhal, Anjali S. Advani, Aziz Nazha, Aaron T. Gerds, Hetty E. Carraway, Mikkael A. Sekeres, Sudipto Mukherjee, Jaroslaw P. Maciejewski, and Tomas Radivoyevitch
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Published
- 2020
- Full Text
- View/download PDF
4. Molecular features of early onset adult myelodysplastic syndrome
- Author
-
Cassandra M. Hirsch, Bartlomiej P. Przychodzen, Tomas Radivoyevitch, Bhumika Patel, Swapna Thota, Michael J. Clemente, Yasunobu Nagata, Thomas LaFramboise, Hetty E. Carraway, Aziz Nazha, Mikkael A. Sekeres, Hideki Makishima, and Jaroslaw P. Maciejewski
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Myelodysplastic syndromes are typically diseases of older adults. Patients in whom the onset is early may have distinct molecular and clinical features or reflect a demographic continuum. The identification of differences between “early onset” patients and those diagnosed at a traditional age has the potential to advance understanding of the pathogenesis of myelodysplasia and may lead to formation of distinct morphological subcategories. We studied a cohort of 634 patients with various subcategories of myelodysplastic syndrome and secondary acute myeloid leukemia, stratifying them based on age at presentation and clinical parameters. We then characterized molecular abnormalities detected by next-generation deep sequencing of 60 genes that are commonly mutated in myeloid malignancies. The number of mutations increased linearly with age and on average, patients >50 years of age had more mutations. TET2, SRSF2, and DNMT3A were more commonly mutated in patients >50 years old compared to patients ≤50 years old. In general, patients >50 years of age also had more mutations in spliceosomal, epigenetic modifier, and RAS gene families. Although there are age-related differences in molecular features among patients with myelodysplasia, most notably in the incidence of SRSF2 mutations, our results suggest that patients ≤50 years old belong to a disease continuum with a distinct pattern of early onset ancestral events.
- Published
- 2017
- Full Text
- View/download PDF
5. Correction: Integrated Multiregional Analysis Proposing a New Model of Colorectal Cancer Evolution.
- Author
-
Ryutaro Uchi, Yusuke Takahashi, Atsushi Niida, Teppei Shimamura, Hidenari Hirata, Keishi Sugimachi, Genta Sawada, Takeshi Iwaya, Junji Kurashige, Yoshiaki Shinden, Tomohiro Iguchi, Hidetoshi Eguchi, Kenichi Chiba, Yuichi Shiraishi, Genta Nagae, Kenichi Yoshida, Yasunobu Nagata, Hiroshi Haeno, Hirofumi Yamamoto, Hideshi Ishii, Yuichiro Doki, Hisae Iinuma, Shin Sasaki, Satoshi Nagayama, Kazutaka Yamada, Shinichi Yachida, Mamoru Kato, Tatsuhiro Shibata, Eiji Oki, Hiroshi Saeki, Ken Shirabe, Yoshinao Oda, Yoshihiko Maehara, Shizuo Komune, Masaki Mori, Yutaka Suzuki, Ken Yamamoto, Hiroyuki Aburatani, Seishi Ogawa, Satoru Miyano, and Koshi Mimori
- Subjects
Genetics ,QH426-470 - Abstract
[This corrects the article DOI: 10.1371/journal.pgen.1005778.].
- Published
- 2017
- Full Text
- View/download PDF
6. Integrated Multiregional Analysis Proposing a New Model of Colorectal Cancer Evolution.
- Author
-
Ryutaro Uchi, Yusuke Takahashi, Atsushi Niida, Teppei Shimamura, Hidenari Hirata, Keishi Sugimachi, Genta Sawada, Takeshi Iwaya, Junji Kurashige, Yoshiaki Shinden, Tomohiro Iguchi, Hidetoshi Eguchi, Kenichi Chiba, Yuichi Shiraishi, Genta Nagae, Kenichi Yoshida, Yasunobu Nagata, Hiroshi Haeno, Hirofumi Yamamoto, Hideshi Ishii, Yuichiro Doki, Hisae Iinuma, Shin Sasaki, Satoshi Nagayama, Kazutaka Yamada, Shinichi Yachida, Mamoru Kato, Tatsuhiro Shibata, Eiji Oki, Hiroshi Saeki, Ken Shirabe, Yoshinao Oda, Yoshihiko Maehara, Shizuo Komune, Masaki Mori, Yutaka Suzuki, Ken Yamamoto, Hiroyuki Aburatani, Seishi Ogawa, Satoru Miyano, and Koshi Mimori
- Subjects
Genetics ,QH426-470 - Abstract
Understanding intratumor heterogeneity is clinically important because it could cause therapeutic failure by fostering evolutionary adaptation. To this end, we profiled the genome and epigenome in multiple regions within each of nine colorectal tumors. Extensive intertumor heterogeneity is observed, from which we inferred the evolutionary history of the tumors. First, clonally shared alterations appeared, in which C>T transitions at CpG site and CpG island hypermethylation were relatively enriched. Correlation between mutation counts and patients' ages suggests that the early-acquired alterations resulted from aging. In the late phase, a parental clone was branched into numerous subclones. Known driver alterations were observed frequently in the early-acquired alterations, but rarely in the late-acquired alterations. Consistently, our computational simulation of the branching evolution suggests that extensive intratumor heterogeneity could be generated by neutral evolution. Collectively, we propose a new model of colorectal cancer evolution, which is useful for understanding and confronting this heterogeneous disease.
- Published
- 2016
- Full Text
- View/download PDF
7. BRCC3 mutations in myeloid neoplasms
- Author
-
Dayong Huang, Yasunobu Nagata, Vera Grossmann, Tomas Radivoyevitch, Yusuke Okuno, Genta Nagae, Naoko Hosono, Susanne Schnittger, Masashi Sanada, Bartlomiej Przychodzen, Ayana Kon, Chantana Polprasert, Wenyi Shen, Michael J. Clemente, James G. Phillips, Tamara Alpermann, Kenichi Yoshida, Niroshan Nadarajah, Mikkael A. Sekeres, Kevin Oakley, Nhu Nguyen, Yuichi Shiraishi, Yusuke Shiozawa, Kenichi Chiba, Hiroko Tanaka, H. Phillip Koeffler, Hans-Ulrich Klein, Martin Dugas, Hiroyuki Aburatani, Satoru Miyano, Claudia Haferlach, Wolfgang Kern, Torsten Haferlach, Yang Du, Seishi Ogawa, and Hideki Makishima
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Next generation sequencing technologies have provided insights into the molecular heterogeneity of various myeloid neoplasms, revealing previously unknown somatic genetic events. In our cohort of 1444 cases analyzed by next generation sequencing, somatic mutations in the gene BRCA1-BRCA2-containing complex 3 (BRCC3) were identified in 28 cases (1.9%). BRCC3 is a member of the JAMM/MPN+ family of zinc metalloproteases capable of cleaving Lys-63 linked polyubiquitin chains, and is implicated in DNA repair. The mutations were located throughout its coding region. The average variant allelic frequency of BRCC3 mutations was 30.1%, and by a serial sample analysis at two different time points a BRCC3 mutation was already identified in the initial stage of a myelodysplastic syndrome. BRCC3 mutations commonly occurred in nonsense (n=12), frameshift (n=4), and splice site (n=5) configurations. Due to the marginal male dominance (odds ratio; 2.00, 0.84–4.73) of BRCC3 mutations, the majority of mutations (n=23; 82%) were hemizygous. Phenotypically, BRCC3 mutations were frequently observed in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms and associated with -Y abnormality (odds ratio; 3.70, 1.25–11.0). Clinically, BRCC3 mutations were also related to higher age (P=0.01), although prognosis was not affected. Knockdown of Brcc3 gene expression in murine bone marrow lineage negative, Sca1 positive, c-kit positive cells resulted in 2-fold more colony formation and modest differentiation defect. Thus, BRCC3 likely plays a role as tumor-associated gene in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms.
- Published
- 2015
- Full Text
- View/download PDF
8. Clonal leukemic evolution in myelodysplastic syndromes with TET2 and IDH1/2 mutations
- Author
-
Tung-Liang Lin, Yasunobu Nagata, Hsiao-Wen Kao, Masashi Sanada, Yusuke Okuno, Chein-Fuang Huang, Der-Cherng Liang, Ming-Chung Kuo, Chang-Liang Lai, En-Hui Lee, Yu-Shu Shih, Hiroko Tanaka, Yuichi Shiraishi, Kenichi Chiba, Tung-Huei Lin, Jin-Hou Wu, Satoru Miyano, Seishi Ogawa, and Lee-Yung Shih
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Somatic mutations of TET2, IDH1, and IDH2 have been described in myelodysplastic syndrome. The impact of these mutations on outcome of myelodysplastic syndrome and their progression to secondary acute myeloid leukemia remains unclear. Mutation status of TET2, IDH1 and IDH2 was investigated in a cohort of 46 paired myelodysplastic syndrome/acute myeloid leukemia samples and 122 non-paired cases with de novo myelodysplastic syndrome, to clarify their roles in the evolution of myelodysplastic syndrome to acute myeloid leukemia. Among the 168 de novo myelodysplastic syndrome patients, the frequency of TET2, IDH1, and IDH2 mutations was 18.5%, 4.2% and 6.0%, respectively. TET2/IDH mutations had no impact on survivals, while TET2 mutations were significantly associated with rapid progression to acute myeloid leukemia. Seventeen of the 46 paired myelodysplastic syndrome/secondary acute myeloid leukemia samples harbored TET2/IDH mutations; none acquired these mutations in acute myeloid leukemia phase. Progression to acute myeloid leukemia was accompanied by evolution of a novel clone or expansion of a minor pre-existing subclone of one or more distinct mutations in 12 of the 17 cases with TET2/IDH mutations. A minor subclone in 3 cases with biallelic TET2 inactivation subsequently expanded, indicating biallelic TET2 mutations play a role in acute myeloid leukemia progression. Twelve patients acquired other genetic lesions, and/or showed increased relative mutant allelic burden of FLT3-ITD, N/K-RAS, CEBPA or RUNX1 during acute myeloid leukemia progression. Our findings provide a novel insight into the role of TET2/IDH mutation in the pathogenesis of myelodysplastic syndrome and subsequent progression to acute myeloid leukemia.
- Published
- 2014
- Full Text
- View/download PDF
9. Epstein-Barr Virus-Related Hemophagocytic Lymphohistiocytosis with Central Nervous System Symptoms
- Author
-
Masahiro, Sakaguchi, Yasunobu, Nagata, Yasuhiro, Terasaki, Atsushi, Takeyoshi, Syunichi, Yasuda, Shunsuke, Honma, Ryosuke, Kinoshita, Atsushi, Marumo, Toshio, Asayama, Shunsuke, Yui, Satoshi, Wakita, Muneo, Okamoto, Yusuke, Kajimoto, Koiti, Inokuchi, and Hiroki, Yamaguchi
- Subjects
General Medicine - Abstract
Hemophagocytic lymphohistiocytosis (HLH) involves pathological histiocytes and phagocytosis of normal blood cells through activation of inflammatory cytokines. We report a case of Epstein-Barr virus-HLH in a 75-year-old woman who presented with fever, thrombocytopenia, and loss of consciousness. Epstein-Barr virus-HLH was diagnosed after we identified massive hemophagocytosis in bone marrow and Epstein-Barr virus DNA in cerebrospinal fluid. The HLH-2004 protocol was applied, and lactate dehydrogenase levels-which reflect HLH disease status-decreased. However, persistent loss of consciousness and multiple organ failure led to the patient' s death on day 18. Most cases of primary and secondary HLH involve pediatric patients; adult cases are rare. Few cases of central nervous system involvement in older adults have been reported. Therefore, accumulation of more data will help in developing better treatment strategies.
- Published
- 2023
10. Data from Amplified EPOR/JAK2 Genes Define a Unique Subtype of Acute Erythroid Leukemia
- Author
-
Seishi Ogawa, Hideki Makishima, Akifumi Takaori-Kondo, Lee-Yung Shih, Felicitas Thol, Michael Heuser, Arnold Ganser, Kazuma Ohyashiki, Takayuki Ishikawa, Jaroslaw P. Maciejewski, Shuichi Miyawaki, Hisashi Tsurumi, Nobuo Sezaki, Kensuke Usuki, Toshiyuki Kitano, Yasushi Miyazaki, Shigeru Chiba, Satoru Miyano, Ken Ishiyama, Hideyuki Nakazawa, Akira Hangaishi, Nobuhiro Hiramoto, Daisuke Morishita, Yasunobu Nagata, Cassandra M. Kerr, Ming-Chung Kuo, Yuichi Shiraishi, Hiroko Tanaka, Kenichi Chiba, Ayana Kon, Takuto Mori, Xingxing Qi, Rurika Okuda, Lanying Zhao, Yotaro Ochi, Ryunosuke Saiki, Akinori Yoda, Yasuhito Nannya, Masahiro M. Nakagawa, Kenichi Yoshida, and June Takeda
- Abstract
Acute erythroid leukemia (AEL) is a unique subtype of acute myeloid leukemia characterized by prominent erythroid proliferation whose molecular basis is poorly understood. To elucidate the underlying mechanism of erythroid proliferation, we analyzed 121 AEL using whole-genome, whole-exome, and/or targeted-capture sequencing, together with transcriptome analysis of 21 AEL samples. Combining publicly available sequencing data, we found a high frequency of gains and amplifications involving EPOR/JAK2 in TP53-mutated cases, particularly those having >80% erythroblasts designated as pure erythroid leukemia (10/13). These cases were frequently accompanied by gains and amplifications of ERG/ETS2 and associated with a very poor prognosis, even compared with other TP53-mutated AEL. In addition to activation of the STAT5 pathway, a common feature across all AEL cases, these AEL cases exhibited enhanced cell proliferation and heme metabolism and often showed high sensitivity to ruxolitinib in vitro and in xenograft models, highlighting a potential role of JAK2 inhibition in therapeutics of AEL.Significance:This study reveals the major role of gains, amplifications, and mutations of EPOR and JAK2 in the pathogenesis of pure erythroleukemia. Their frequent response to ruxolitinib in patient-derived xenograft and cell culture models highlights a possible therapeutic role of JAK2 inhibition for erythroleukemia with EPOR/JAK2-involving lesions.This article is highlighted in the In This Issue feature, p. 369
- Published
- 2023
11. Supplementary Figure from Amplified EPOR/JAK2 Genes Define a Unique Subtype of Acute Erythroid Leukemia
- Author
-
Seishi Ogawa, Hideki Makishima, Akifumi Takaori-Kondo, Lee-Yung Shih, Felicitas Thol, Michael Heuser, Arnold Ganser, Kazuma Ohyashiki, Takayuki Ishikawa, Jaroslaw P. Maciejewski, Shuichi Miyawaki, Hisashi Tsurumi, Nobuo Sezaki, Kensuke Usuki, Toshiyuki Kitano, Yasushi Miyazaki, Shigeru Chiba, Satoru Miyano, Ken Ishiyama, Hideyuki Nakazawa, Akira Hangaishi, Nobuhiro Hiramoto, Daisuke Morishita, Yasunobu Nagata, Cassandra M. Kerr, Ming-Chung Kuo, Yuichi Shiraishi, Hiroko Tanaka, Kenichi Chiba, Ayana Kon, Takuto Mori, Xingxing Qi, Rurika Okuda, Lanying Zhao, Yotaro Ochi, Ryunosuke Saiki, Akinori Yoda, Yasuhito Nannya, Masahiro M. Nakagawa, Kenichi Yoshida, and June Takeda
- Abstract
Supplementary Figure from Amplified EPOR/JAK2 Genes Define a Unique Subtype of Acute Erythroid Leukemia
- Published
- 2023
12. Supplementary Table from Amplified EPOR/JAK2 Genes Define a Unique Subtype of Acute Erythroid Leukemia
- Author
-
Seishi Ogawa, Hideki Makishima, Akifumi Takaori-Kondo, Lee-Yung Shih, Felicitas Thol, Michael Heuser, Arnold Ganser, Kazuma Ohyashiki, Takayuki Ishikawa, Jaroslaw P. Maciejewski, Shuichi Miyawaki, Hisashi Tsurumi, Nobuo Sezaki, Kensuke Usuki, Toshiyuki Kitano, Yasushi Miyazaki, Shigeru Chiba, Satoru Miyano, Ken Ishiyama, Hideyuki Nakazawa, Akira Hangaishi, Nobuhiro Hiramoto, Daisuke Morishita, Yasunobu Nagata, Cassandra M. Kerr, Ming-Chung Kuo, Yuichi Shiraishi, Hiroko Tanaka, Kenichi Chiba, Ayana Kon, Takuto Mori, Xingxing Qi, Rurika Okuda, Lanying Zhao, Yotaro Ochi, Ryunosuke Saiki, Akinori Yoda, Yasuhito Nannya, Masahiro M. Nakagawa, Kenichi Yoshida, and June Takeda
- Abstract
Supplementary Table from Amplified EPOR/JAK2 Genes Define a Unique Subtype of Acute Erythroid Leukemia
- Published
- 2023
13. Supplementary Fig 5 from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Supplementary Fig. 5. Kaplan-Meier analysis of the survival of patients with different mutations.
- Published
- 2023
14. Supplementary Figure legends from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Legends for Supplementary Figures
- Published
- 2023
15. Supplements from IDH1/2 Mutations Sensitize Acute Myeloid Leukemia to PARP Inhibition and This Is Reversed by IDH1/2-Mutant Inhibitors
- Author
-
Jaroslaw P. Maciejewski, Cornelis J.F. van Noorden, Mikkael A. Sekeres, Sudipto Mukherjee, Hetty E. Carraway, Johanna W. Wilmink, Fonnet E. Bleeker, Mingjiang Xu, Hideki Makishima, Bartolomiej Przychodzen, Mohammed Khurshed, Yasunobu Nagata, Tomas Radivoyevitch, and Remco J. Molenaar
- Abstract
Supplementary Figures, Methods and Tables
- Published
- 2023
16. Data from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Current standard of care for patients with pediatric acute lymphoblastic leukemia (ALL) is mainly effective, with high remission rates after treatment. However, the genetic perturbations that give rise to this disease remain largely undefined, limiting the ability to address resistant tumors or develop less toxic targeted therapies. Here, we report the use of next-generation sequencing to interrogate the genetic and pathogenic mechanisms of 240 pediatric ALL cases with their matched remission samples. Commonly mutated genes fell into several categories, including RAS/receptor tyrosine kinases, epigenetic regulators, transcription factors involved in lineage commitment, and the p53/cell-cycle pathway. Unique recurrent mutational hotspots were observed in epigenetic regulators CREBBP (R1446C/H), WHSC1 (E1099K), and the tyrosine kinase FLT3 (K663R, N676K). The mutant WHSC1 was established as a gain-of-function oncogene, while the epigenetic regulator ARID1A and transcription factor CTCF were functionally identified as potential tumor suppressors. Analysis of 28 diagnosis/relapse trio patients plus 10 relapse cases revealed four evolutionary paths and uncovered the ordering of acquisition of mutations in these patients. This study provides a detailed mutational portrait of pediatric ALL and gives insights into the molecular pathogenesis of this disease. Cancer Res; 77(2); 390–400. ©2016 AACR.
- Published
- 2023
17. Supplementary Fig 4 from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Supplementary Fig. 4. p53 pathway is dysregulated in ALL.
- Published
- 2023
18. Supplementary Fig 2 from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Supplementary Fig. 2. Mutations found in the ASXL family of genes.
- Published
- 2023
19. Data from IDH1/2 Mutations Sensitize Acute Myeloid Leukemia to PARP Inhibition and This Is Reversed by IDH1/2-Mutant Inhibitors
- Author
-
Jaroslaw P. Maciejewski, Cornelis J.F. van Noorden, Mikkael A. Sekeres, Sudipto Mukherjee, Hetty E. Carraway, Johanna W. Wilmink, Fonnet E. Bleeker, Mingjiang Xu, Hideki Makishima, Bartolomiej Przychodzen, Mohammed Khurshed, Yasunobu Nagata, Tomas Radivoyevitch, and Remco J. Molenaar
- Abstract
Purpose: Somatic mutations in IDH1/2 occur in approximately 20% of patients with myeloid neoplasms, including acute myeloid leukemia (AML). IDH1/2MUT enzymes produce D-2-hydroxyglutarate (D2HG), which associates with increased DNA damage and improved responses to chemo/radiotherapy and PARP inhibitors in solid tumor cells. Whether this also holds true for IDH1/2MUT AML is not known.Experimental Design: Well-characterized primary IDH1MUT, IDH2MUT, and IDH1/2WT AML cells were analyzed for DNA damage and responses to daunorubicin, ionizing radiation, and PARP inhibitors.Results: IDH1/2MUT caused increased DNA damage and sensitization to daunorubicin, irradiation, and the PARP inhibitors olaparib and talazoparib in AML cells. IDH1/2MUT inhibitors protected against these treatments. Combined treatment with a PARP inhibitor and daunorubicin had an additive effect on the killing of IDH1/2MUT AML cells. We provide evidence that the therapy sensitivity of IDH1/2MUT cells was caused by D2HG-mediated downregulation of expression of the DNA damage response gene ATM and not by altered redox responses due to metabolic alterations in IDH1/2MUT cells.Conclusions: IDH1/2MUT AML cells are sensitive to PARP inhibitors as monotherapy but especially when combined with a DNA-damaging agent, such as daunorubicin, whereas concomitant administration of IDH1/2MUT inhibitors during cytotoxic therapy decrease the efficacy of both agents in IDH1/2MUT AML. These results advocate in favor of clinical trials of PARP inhibitors either or not in combination with daunorubicin in IDH1/2MUT AML. Clin Cancer Res; 24(7); 1705–15. ©2018 AACR.
- Published
- 2023
20. Supplementary Fig 1 from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Supplementary Fig. 1. Mutational spectrum of pediatric ALL.
- Published
- 2023
21. Supplementary Tables from Mutational Landscape of Pediatric Acute Lymphoblastic Leukemia
- Author
-
H. Phillip Koeffler, Henry Yang, Lee-Yung Shih, Seishi Ogawa, Der-Cherng Liang, Satoru Miyano, Steven M. Kornblau, Hagop M. Kantarjian, Michael Lill, Hema Preethi, Lucia Torres Fernández, Masashi Sanada, Vikas Madan, Li-Zhen Liu, Su-Lin Lim, Liang Xu, Yan-Yi Jiang, Manoj Garg, De-Chen Lin, Xin-Yi Loh, Jin-Fen Xiao, Yasunobu Nagata, Norihiko Kawamata, Allen Eng Juh Yeoh, Anand Mayakonda Thippeswamy, Wenwen Chien, Kar-Tong Tan, Qiao-Yang Sun, and Ling-Wen Ding
- Abstract
Supplementary Table 1. Samples used for whole exome and targeted sequencing. Supplementary Table 2. Clinical information of patients. Supplementary Table 3. 560 Genes screened in the targeted sequencing. Supplementary Table 4. Sequences of shRNA or CRISPR-Cas9 sgRNA used in this study. Supplementary Table 5. Sequences of real-time PCR primers used in this study. Supplementary Table 6. Sequences of primers used for Sanger validation of CRISPR-Cas9 indel. Supplementary Table 7. Mutations of KRAS identified in this study have been found in a variety of cancers. Supplementary Table 8. Mutations of PTPN11 in other cancers occurring in the same location as in our ALL cohort. Supplementary Table 9. Recurrent mutation sites of FLT3 in different cancers. Supplementary Table 10. Cell lines or cancer samples harboring either E1099K or T1150A hotspot mutation of WHSC1. Supplementary Table 11. R1446 mutations of CREBBP recurrently occurred in a variety of cancers. Supplementary Table 12. Mutational hotspot (D1399) of EP300 occurring in other cancers occurring in the same location as in our ALL cohort.Supplementary Table 13. Rare mutations occurring in our ALL cohort (mutated in one individual each), but these mutations have been recurrently documented in the COSMIC cancer mutations database. Supplementary Table 14. Mutations of genes involved in DNA repair pathway in relapse samples.
- Published
- 2023
22. Germline Risks and Clinical Impacts of DDX41 Mutations in Myeloid Malignancies
- Author
-
Hideki Makishima, Ryunosuke Saiki, Yasuhito Nannya, Sophia C. Korotev, Carmelo Gurnari, June Takeda, Yukihide Momozawa, Steve Best, Pramila Krishnamurthy, Tetsuichi Yoshizato, Yoshiko Atsuta, Yusuke Shiozawa, Yuka Iijima-Yamashita, Kenichi Yoshida, Yuichi Shiraishi, Yasunobu Nagata, Nobuyuki Kakiuchi, Makoto Onizuka, Kenichi Chiba, Hiroko Tanaka, Ayana Kon, Yotaro Ochi, Masahiro Marshall Nakagawa, Rurika Okuda, Takuto Mori, Akinori Yoda, Hidehiro Itonaga, Yasushi Miyazaki, Masashi Sanada, Takayuki Ishikawa, Shigeru Chiba, Hisashi Tsurumi, Senji Kasahara, Carsten Müller-Tidow, Akifumi Takaori-Kondo, Kazuma Ohyashiki, Toru Kiguchi, Fumihiko Matsuda, Joop H. Jansen, Chantana Polprasert, Piers Blombery, Yoichiro Kamatani, Satoru Miyano, Luca Malcovati, Torsten Haferlach, Michiaki Kubo, Mario Cazzola, Austin Kulasekararaj, Lucy A. Godley, Jaroslaw P. Maciejewski, and Seishi Ogawa
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
23. Safety and efficacy of high‐dose cytarabine MEAM therapy and other treatments for auto‐peripheral blood stem cell transplantation: A retrospective comparative study
- Author
-
Shunsuke Yui, Satoshi Wakita, Yasunobu Nagata, Yasuko Kuribayashi, Toshio Asayama, Yusuke Fujiwara, Masahiro Sakaguchi, Satoshi Yamanaka, Atsushi Marumo, Ikuko Omori, Ryosuke Kinoshita, Daishi Onai, Mika Sunakawa, Yuta Kaito, Kazuki Inai, Taichiro Tokura, Atsushi Takeyoshi, Shunichi Yasuda, Shunsuke Honma, Kazutaka Nakayama, Tsuneaki Hirakawa, Kunihito Arai, Tomoaki Kitano, Muneo Okamoto, Koiti Inokuchi, and Hiroki Yamaguchi
- Subjects
Oncology ,General Medicine - Abstract
The MEAM regimen consisting of ranimustine (MCNU), etoposide (ETP), cytarabine (Ara-C), and melphalan (MEL) is widely used before auto-peripheral blood stem cell transplantation (auto-PBSCT) for malignant lymphoma in Japan. The MEAM regimen generally consists of 200-400 mg/mThe high-dose Ara-C MEAM protocol consisted of MCNU 300 mg/mAll patients got engraftment and there were no cases of treatment-related mortality. In all cases, the 3-year overall survival (OS) and progression-free survival (PFS) after transplantation were 80.6% and 65.7%, respectively. Twenty-one cases of diffuse large B-cell lymphoma recurrence, for which there is proven usefulness of auto-PBSCT, showed good results after transplantation, with the 3-year OS and PFS after transplantation being 100% and 74.3%, respectively.The safety and efficacy of high-dose Ara-C MEAM therapy were demonstrated, but the expected therapeutic effect on central nervous system lesions could not be fully evaluated owing to the small number of cases.
- Published
- 2022
24. Abstract TP202: Sex-specific Differences In Risk Profiles For Cancer Among 19702 Japanese Patients With Ischemic Stroke: The Biobank Japan Project
- Author
-
Takashi Shimoyama, Koichi Matsuda, Yoichiro Kamatani, Yasunobu Nagata, Hiroki Yamaguchi, and Kazumi Kimura
- Subjects
Advanced and Specialized Nursing ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine - Abstract
Background: There is limited data on sex-specific differences in subtypes and risk profiles of cancer among ischemic stroke patients. Methods: Among the subjects registered in the BioBank Japan database, 19702 ischemic stroke patients (n= 12241 male and n= 7261 female, median age 71 years old) were analyzed. We compared the prevalence of 20 cancer types between men and women. The multivariate logistic analysis was used to explore the association between risk factors and cancer by sex. Results: Any cancer was present in 1656 (8.4%) subsets of the overall cohort. The prevalence was highest for colorectal cancer (1.9%) followed by gastric cancer (1.6%), prostate cancer (1.2%), breast cancer (0.8%), and lung cancer (0.5%). Incidence was significantly higher in men than women for any (8.7% vs. 7.8%, P=0.029), colorectal (2.1% vs. 1.5%, P=0.003), gastric (2.0% vs. 1.0%, P Conclusion: Men have a higher prevalence of cancer including shared 6 anatomic sites, while sex-specific cancer was more common in women. Male predominance is largely explained by risk profiles including older age, chronic renal failure, atrial fibrillation or flutter, and smoking. These results indicate that cancer among stroke patients shares common risk factors for stroke or cardiovascular disease in men but not women.
- Published
- 2023
25. Germ line DDX41 mutations define a unique subtype of myeloid neoplasms
- Author
-
Hideki Makishima, Ryunosuke Saiki, Yasuhito Nannya, Sophia Korotev, Carmelo Gurnari, June Takeda, Yukihide Momozawa, Steve Best, Pramila Krishnamurthy, Tetsuichi Yoshizato, Yoshiko Atsuta, Yusuke Shiozawa, Yuka Iijima-Yamashita, Kenichi Yoshida, Yuichi Shiraishi, Yasunobu Nagata, Nobuyuki Kakiuchi, Makoto Onizuka, Kenichi Chiba, Hiroko Tanaka, Ayana Kon, Yotaro Ochi, Masahiro M. Nakagawa, Rurika Okuda, Takuto Mori, Akinori Yoda, Hidehiro Itonaga, Yasushi Miyazaki, Masashi Sanada, Takayuki Ishikawa, Shigeru Chiba, Hisashi Tsurumi, Senji Kasahara, Carsten Müller-Tidow, Akifumi Takaori-Kondo, Kazuma Ohyashiki, Toru Kiguchi, Fumihiko Matsuda, Joop H. Jansen, Chantana Polprasert, Piers Blombery, Yoichiro Kamatani, Satoru Miyano, Luca Malcovati, Torsten Haferlach, Michiaki Kubo, Mario Cazzola, Austin G. Kulasekararaj, Lucy A. Godley, Jaroslaw P. Maciejewski, and Seishi Ogawa
- Subjects
All institutes and research themes of the Radboud University Medical Center ,Cancer development and immune defence Radboud Institute for Molecular Life Sciences [Radboudumc 2] ,Immunology ,Cell Biology ,Hematology ,Settore MED/15 ,Biochemistry - Abstract
Germ line DDX41 variants have been implicated in late-onset myeloid neoplasms (MNs). Despite an increasing number of publications, many important features of DDX41-mutated MNs remain to be elucidated. Here we performed a comprehensive characterization of DDX41-mutated MNs, enrolling a total of 346 patients with DDX41 pathogenic/likely-pathogenic (P/LP) germ line variants and/or somatic mutations from 9082 MN patients, together with 525 first-degree relatives of DDX41-mutated and wild-type (WT) patients. P/LP DDX41 germ line variants explained ∼80% of known germ line predisposition to MNs in adults. These risk variants were 10-fold more enriched in Japanese MN cases (n = 4461) compared with the general population of Japan (n = 20 238). This enrichment of DDX41 risk alleles was much more prominent in male than female (20.7 vs 5.0). P/LP DDX41 variants conferred a large risk of developing MNs, which was negligible until 40 years of age but rapidly increased to 49% by 90 years of age. Patients with myelodysplastic syndromes (MDS) along with a DDX41-mutation rapidly progressed to acute myeloid leukemia (AML), which was however, confined to those having truncating variants. Comutation patterns at diagnosis and at progression to AML were substantially different between DDX41-mutated and WT cases, in which none of the comutations affected clinical outcomes. Even TP53 mutations made no exceptions and their dismal effect, including multihit allelic status, on survival was almost completely mitigated by the presence of DDX41 mutations. Finally, outcomes were not affected by the conventional risk stratifications including the revised/molecular International Prognostic Scoring System. Our findings establish that MDS with DDX41-mutation defines a unique subtype of MNs that is distinct from other MNs.
- Published
- 2023
26. Cooperative Effects of SRSF2 and STAG2 mutations on Development of Myelodysplastic Syndrome and Its Related Disorders
- Author
-
Takuto Mori, Yotaro Ochi, Ayana Kon, Tetsuichi Yoshizato, Haruhiko Koseki, Manabu Nakayama, Akinori Yoda, Masahiro Marshall Nakagawa, June Takeda, Ryunosuke Saiki, Rurika Okuda, Yasunobu Nagata, Kenichi Yoshida, Masashi Sanada, Hideki Makishima, Akifumi Takaori-Kondo, Jaroslaw P. Maciejewski, Yoshiko Atsuta, Torsten Haferlach, Yasuhito Nannya, and Seishi Ogawa
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
27. Machine learning demonstrates that somatic mutations imprint invariant morphologic features in myelodysplastic syndromes
- Author
-
Hassan Awada, Brian P. Hobbs, Jacob G. Scott, Sunisa Kongkiatkamon, Aziz Nazha, Jaroslaw P. Maciejewski, Tomas Radivoyevitch, Yasunobu Nagata, Inom Mirzaev, Ran Zhao, Mikkael A. Sekeres, Hideki Makishima, and Cassandra M Kerr
- Subjects
Adult ,Male ,Somatic cell ,Immunology ,Erythroid dysplasia ,Computational biology ,Biology ,Biochemistry ,Machine Learning ,Genetic signature ,Monocytosis ,Pathognomonic ,Genotype ,medicine ,Humans ,Genetic Association Studies ,Aged ,Myelodysplastic syndromes ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Pancytopenia ,Myelodysplastic Syndromes ,Mutation ,Female - Abstract
Morphologic interpretation is the standard in diagnosing myelodysplastic syndrome (MDS), but it has limitations, such as varying reliability in pathologic evaluation and lack of integration with genetic data. Somatic events shape morphologic features, but the complexity of morphologic and genetic changes makes clear associations challenging. This article interrogates novel clinical subtypes of MDS using a machine-learning technique devised to identify patterns of cooccurrence among morphologic features and genomic events. We sequenced 1079 MDS patients and analyzed bone marrow morphologic alterations and other clinical features. A total of 1929 somatic mutations were identified. Five distinct morphologic profiles with unique clinical characteristics were defined. Seventy-seven percent of higher-risk patients clustered in profile 1. All lower-risk (LR) patients clustered into the remaining 4 profiles: profile 2 was characterized by pancytopenia, profile 3 by monocytosis, profile 4 by elevated megakaryocytes, and profile 5 by erythroid dysplasia. These profiles could also separate patients with different prognoses. LR MDS patients were classified into 8 genetic signatures (eg, signature A had TET2 mutations, signature B had both TET2 and SRSF2 mutations, and signature G had SF3B1 mutations), demonstrating association with specific morphologic profiles. Six morphologic profiles/genetic signature associations were confirmed in a separate analysis of an independent cohort. Our study demonstrates that nonrandom or even pathognomonic relationships between morphology and genotype to define clinical features can be identified. This is the first comprehensive implementation of machine-learning algorithms to elucidate potential intrinsic interdependencies among genetic lesions, morphologies, and clinical prognostic in attributes of MDS.
- Published
- 2020
28. Analysis of distinct SF3B1 hotspot mutations in relation to clinical phenotypes and response to therapy in myeloid neoplasia
- Author
-
Yazan F. Madanat, Teodora Kuzmanovic, Sudipto Mukherjee, Heesun J. Rogers, Anjali S. Advani, Aaron T. Gerds, Bhumika J. Patel, Yasunobu Nagata, Mikkael A. Sekeres, Yogen Saunthararajah, Cassandra M Kerr, Aziz Nazha, Jennifer S. Carew, Steffan T. Nawrocki, Hetty E. Carraway, Francesc Solé, Valeria Visconte, Ying Ni, Jack Khouri, Hassan Awada, Vera Adema, and Jaroslaw P. Maciejewski
- Subjects
Genetics ,chemistry.chemical_classification ,Cancer Research ,Response to therapy ,business.industry ,Hematology ,Biology ,Phenotype ,Amino acid ,Myeloid neoplasia ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,business ,030215 immunology - Abstract
Here we investigate single codon SF3B1 mutations (K700E, K666, H662) to identify whether discrete amino acid substitutions may influence clinical phenotypes, survival and response to treatments in ...
- Published
- 2020
29. Rare germline variant contributions to myeloid malignancy susceptibility
- Author
-
Teodora Kuzmanovic, Cassandra M Kerr, Francesc Solé, Samuel T Li, Vera Adema, Ruipeng Wei, Bartlomiej P Przychodzen, Yasunobu Nagata, Ruqi Shi, Thomas LaFramboise, Jaroslaw P. Maciejewski, and Janet Wang
- Subjects
Myeloid Malignancy ,Cancer Research ,Myeloproliferative Disorders ,Extramural ,business.industry ,MEDLINE ,Hematology ,Article ,Germline ,Text mining ,Oncology ,Myelodysplastic Syndromes ,Cancer research ,Humans ,Medicine ,Genetic Predisposition to Disease ,business ,Germ-Line Mutation - Published
- 2020
30. Amplified EPOR/JAK2 Genes Define a Unique Subtype of Acute Erythroid Leukemia
- Author
-
June Takeda, Kenichi Yoshida, Masahiro M. Nakagawa, Yasuhito Nannya, Akinori Yoda, Ryunosuke Saiki, Yotaro Ochi, Lanying Zhao, Rurika Okuda, Xingxing Qi, Takuto Mori, Ayana Kon, Kenichi Chiba, Hiroko Tanaka, Yuichi Shiraishi, Ming-Chung Kuo, Cassandra M. Kerr, Yasunobu Nagata, Daisuke Morishita, Nobuhiro Hiramoto, Akira Hangaishi, Hideyuki Nakazawa, Ken Ishiyama, Satoru Miyano, Shigeru Chiba, Yasushi Miyazaki, Toshiyuki Kitano, Kensuke Usuki, Nobuo Sezaki, Hisashi Tsurumi, Shuichi Miyawaki, Jaroslaw P. Maciejewski, Takayuki Ishikawa, Kazuma Ohyashiki, Arnold Ganser, Michael Heuser, Felicitas Thol, Lee-Yung Shih, Akifumi Takaori-Kondo, Hideki Makishima, and Seishi Ogawa
- Subjects
Leukemia, Myeloid, Acute ,Mutation ,Receptors, Erythropoietin ,Humans ,Exome ,General Medicine ,Leukemia, Erythroblastic, Acute ,Janus Kinase 2 ,Prognosis - Abstract
Acute erythroid leukemia (AEL) is a unique subtype of acute myeloid leukemia characterized by prominent erythroid proliferation whose molecular basis is poorly understood. To elucidate the underlying mechanism of erythroid proliferation, we analyzed 121 AEL using whole-genome/exome and/or targeted-capture sequencing, together with transcriptome analysis of 21 AEL samples. Combining publicly available sequencing data, we found a high frequency of gains/amplifications involving EPOR/JAK2 in TP53-mutated cases, particularly those having >80% erythroblasts designated as pure erythroid leukemia (10/13). These cases were frequently accompanied by gains/amplifications of ERG/ETS2 and associated with a very poor prognosis, even compared with other TP53-mutated AEL. In addition to activation of the STAT5 pathway, a common feature across all AEL cases, these AEL cases exhibited enhanced cell proliferation and heme metabolism and often showed high sensitivity to ruxolitinib in vitro and in xenograft models, highlighting a potential role of JAK2 inhibition in therapeutics of AEL., ゲノム解析から急性赤白血病の変異プロファイルと治療標的を解明 --特定の遺伝子変異群の組み合わせと、特徴となる遺伝子の増幅が鍵--. 京都大学プレスリリース. 2022-08-05.
- Published
- 2021
31. Invariant patterns of clonal succession determine specific clinical features of myelodysplastic syndromes
- Author
-
Mohammad Fahad B Asad, Tomas Radivoyevitch, Yuichi Shiraishi, Satoru Miyano, Jaroslaw P. Maciejewski, Hassan Awada, Abhinav Goyal, Hiroko Tanaka, Kenichi Yoshida, Torsten Haferlach, Hiromichi Suzuki, Sudipto Mukherjee, Aziz Nazha, Kenichi Chiba, Bartlomiej P Przychodzen, Yasunobu Nagata, Thomas LaFramboise, Hideki Makishima, Seishi Ogawa, Mikkael A. Sekeres, Mai Aly, Teodora Kuzmanovic, Tetsuichi Yoshizato, and Cassandra M Kerr
- Subjects
Male ,0301 basic medicine ,Myeloid ,Somatic cell ,Science ,General Physics and Astronomy ,Biology ,medicine.disease_cause ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,Exome Sequencing ,Cancer genomics ,medicine ,Humans ,lcsh:Science ,Exome sequencing ,Aged ,Genetics ,Multidisciplinary ,Myelodysplastic syndromes ,Clonal architecture ,General Chemistry ,Phosphoproteins ,Splicing Factor U2AF ,medicine.disease ,Phenotype ,Hematopoiesis ,Leukemia, Myeloid, Acute ,Leukemia ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Primary Myelofibrosis ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Mutation ,Female ,lcsh:Q ,RNA Splicing Factors ,KRAS ,Tumor Suppressor Protein p53 ,Myelodysplastic syndrome - Abstract
Myelodysplastic syndromes (MDS) arise in older adults through stepwise acquisitions of multiple somatic mutations. Here, analyzing 1809 MDS patients, we infer clonal architecture by using a stringent, the single-cell sequencing validated PyClone bioanalytic pipeline, and assess the position of the mutations within the clonal architecture. All 3,971 mutations are grouped based on their rank in the deduced clonal hierarchy (dominant and secondary). We evaluated how they affect the resultant morphology, progression, survival and response to therapies. Mutations of SF3B1, U2AF1, and TP53 are more likely to be dominant, those of ASXL1, CBL, and KRAS are secondary. Among distinct combinations of dominant/secondary mutations we identified 37 significant relationships, of which 12 affect clinical phenotypes, 5 cooperatively associate with poor prognosis. They also predict response to hypomethylating therapies. The clonal hierarchy has distinct ranking and the resultant invariant combinations of dominant/secondary mutations yield novel insights into the specific clinical phenotype of MDS., Stepwise acquisition of mutations gives rise to myelodysplastic syndrome (MDS) in older adults. Here, the authors infer the clonal hierarchy of 1809 MDS patients, revealing insights into the evolution of dominant/secondary mutations and how these impact clinical phenotypes like leukemic progression and therapy response.
- Published
- 2019
32. Large granular lymphocytic leukemia coexists with myeloid clones and myelodysplastic syndrome
- Author
-
Sudipto Mukherjee, Hetty E. Carraway, Vera Adema, Alan E. Lichtin, Bhumika J. Patel, Teodora Kuzmanovic, Yasunobu Nagata, Ashwin Kishtagari, Jaroslaw P. Maciejewski, Sanghee Hong, Yogen Saunthararajah, Aziz Nazha, Valeria Visconte, Hassan Awada, Mikkael A. Sekeres, Cassandra M Kerr, and Jibran Durrani
- Subjects
Aged, 80 and over ,Male ,Cancer Research ,Myeloid ,business.industry ,Extramural ,Cell growth ,T-Lymphocytes ,Large granular lymphocytic leukemia ,Hematology ,Middle Aged ,medicine.disease ,Leukemia, Large Granular Lymphocytic ,Leukemia ,medicine.anatomical_structure ,Oncology ,Myelodysplastic Syndromes ,Cancer research ,Humans ,Medicine ,Female ,Myeloid Cells ,business ,Aged ,Cell Proliferation - Published
- 2019
33. Leukemia evolving from paroxysmal nocturnal hemoglobinuria
- Author
-
Jaroslaw P. Maciejewski, Shafia Rahman, Bhumika J. Patel, Hetty E. Carraway, Mohammad Fahad B Asad, Yasunobu Nagata, Mikkael A. Sekeres, Valeria Visconte, Ashwin Kishtagari, Vera Adema, Sunisa Kongkiatkamon, Christina Snider, Hassan Awada, Cassandra M Kerr, Amy C Graham, and Jibran Durrani
- Subjects
Adult ,Aged, 80 and over ,Male ,Cancer Research ,Leukemia ,business.industry ,Hemoglobinuria, Paroxysmal ,Hematology ,Middle Aged ,medicine.disease ,Article ,Young Adult ,Cell Transformation, Neoplastic ,Oncology ,Immunology ,Paroxysmal nocturnal hemoglobinuria ,medicine ,Humans ,Female ,Cancer epigenetics ,business ,Aged - Published
- 2019
34. Genomics of therapy-related myeloid neoplasms
- Author
-
Teodora Kuzmanovic, Jaroslaw P. Maciejewski, Deepak Singhal, Yasunobu Nagata, Devendra K Hiwase, Aziz Nazha, Hetty E. Carraway, Bhumika J. Patel, Srinivasa R. Sanikommu, Aaron T. Gerds, Mikkael A. Sekeres, Hassan Awada, Anjali S. Advani, Sudipto Mukherjee, Tomas Radivoyevitch, Cassandra M Kerr, Bartlomiej P Przychodzen, and Babal K. Jha
- Subjects
Oncology ,medicine.medical_specialty ,Myeloid ,Therapy related ,business.industry ,MEDLINE ,Neoplasms, Second Primary ,Genomics ,Hematology ,Second primary cancer ,medicine.disease ,Leukemia, Myeloid, Acute ,Leukemia ,medicine.anatomical_structure ,Neoplasms ,Internal medicine ,medicine ,Humans ,Online Only Articles ,business - Published
- 2019
35. Distinct clinical and biological implications of CUX1 in myeloid neoplasms
- Author
-
Valeria Visconte, Bartlomiej P Przychodzen, Mai Aly, Alain Nepveu, Hideki Makishima, Vera Adema, Yasunobu Nagata, Jaroslaw P. Maciejewski, Teodora Kuzmanovic, Cassandra M Kerr, Naoko Hosono, Aziz Nazha, Mikkael A. Sekeres, Mohamed E. Abazeed, Zubaidah M. Ramdzan, and Suresh Kumar Balasubramanian
- Subjects
Myeloid ,DNA repair ,Somatic cell ,Myelodysplastic syndromes ,Myeloid leukemia ,Locus (genetics) ,Hematology ,Biology ,medicine.disease ,Frameshift mutation ,Loss of heterozygosity ,medicine.anatomical_structure ,medicine ,Cancer research - Abstract
Somatic mutations of the CUT-like homeobox 1 ( CUX1 ) gene ( CUX1 MT ) can be found in myeloid neoplasms (MNs), in particular, in myelodysplastic syndromes (MDSs). The CUX1 locus is also deleted in 3 of 4 MN cases with −7/del(7q). A cohort of 1480 MN patients was used to characterize clinical features and clonal hierarchy associated with CUX1 MT and CUX1 deletions ( CUX1 DEL ) and to analyze their functional consequences in vitro. CUX1 MT were present in 4% of chronic MNs. CUX1 DEL were preferentially found in advanced cases (6%). Most MDS and acute myeloid leukemia (AML) patients with −7/del(7q) and up to 15% of MDS patients and 5% of AML patients diploid for the CUX1 locus exhibited downmodulated CUX1 expression. In 75% of mutant cases, CUX1 MT were heterozygous, whereas microdeletions and homozygous and compound-heterozygous mutations were less common. CUX MT/DEL were associated with worse survival compared with CUX1 WT . Within the clonal hierarchy, 1 of 3 CUX1 MT served as founder events often followed by secondary BCOR and ASXL1 subclonal hits, whereas TET2 was the most common ancestral lesion, followed by subclonal CUX1 MT . Comet assay of patients’ bone marrow progenitor cells and leukemic cell lines performed in various experimental conditions revealed that frameshift mutations, hemizygous deletions, or experimental CUX1 knockdown decrease the repair of oxidized bases. These functional findings may explain why samples with either CUX1 MT or low CUX1 expression coincided with significantly higher numbers of somatic hits by whole-exome sequencing. Our findings implicate the DNA repair dysfunction resulting from CUX1 lesions in the pathogenesis of MNs, in which they lead to a mutator phenotype.
- Published
- 2019
36. Molecular heterogeneity in peripheral T-cell lymphoma, not otherwise specified revealed by comprehensive genetic profiling
- Author
-
Tetsuichi Yoshizato, Yuichi Shiraishi, Nobuyuki Kakiuchi, Kengo Takeuchi, Ryota Matsuoka, Hiroko Tanaka, Yusuke Kito, Kazuya Shimoda, Tadashi Yoshino, Kenji Nishida, Takayuki Ishikawa, Kenichi Chiba, Kana Sakamoto, Lanying Zhao, Yusuke Shiozawa, Hiroaki Miyoshi, Hiroo Ueno, Nobuhiro Hiramoto, Kenichi Yoshida, Koichi Ohshima, Junichi Kitagawa, Shigeru Chiba, Yasuhito Nanya, Hisashi Tsurumi, Masashi Sanada, Nobuhiko Nakamura, Yasunobu Nagata, Yasuhide Takeuchi, Satoru Miyano, Hideki Makishima, Masayuki Noguchi, June Takeda, Keisuke Kataoka, Tatsuhiko Miyazaki, Yasuharu Sato, Mamiko Sakata-Yanagimoto, Masahiro Nakagawa, Yotaro Ochi, Yasunori Kogure, Seishi Ogawa, Yosaku Watatani, and Yuka Gion
- Subjects
Male ,0301 basic medicine ,Genome instability ,Cancer Research ,DNA Copy Number Variations ,Peripheral T-cell lymphoma not otherwise specified ,Genomics ,Computational biology ,Biology ,Genomic Instability ,Genetic Heterogeneity ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,Exome Sequencing ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Alleles ,Genetic Association Studies ,Exome sequencing ,Genetic heterogeneity ,Gene Expression Profiling ,Computational Biology ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Lymphoma, T-Cell, Peripheral ,Hematology ,medicine.disease ,Gene expression profiling ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Female ,Tumor Escape ,Signal Transduction - Abstract
Peripheral T-cell lymphoma, not otherwise specified (PTCL, NOS) is a diagnosis of exclusion, being the most common entity in mature T-cell neoplasms, and its molecular pathogenesis remains significantly understudied. Here, combining whole-exome and targeted-capture sequencing, gene-expression profiling, and immunohistochemical analysis of tumor samples from 133 cases, we have delineated the entire landscape of somatic alterations, and discovered frequently affected driver pathways in PTCL, NOS, with and without a T-follicular helper (TFH) cell phenotype. In addition to previously reported mutational targets, we identified a number of novel recurrently altered genes, such as KMT2C, SETD1B, YTHDF2, and PDCD1. We integrated these genetic drivers using hierarchical clustering and identified a previously undescribed molecular subtype characterized by TP53 and/or CDKN2A mutations and deletions in non-TFH PTCL, NOS. This subtype exhibited different prognosis and unique genetic features associated with extensive chromosomal instability, which preferentially affected molecules involved in immune escape and transcriptional regulation, such as HLA-A/B and IKZF2. Taken together, our findings provide novel insights into the molecular pathogenesis of PTCL, NOS by highlighting their genetic heterogeneity. These results should help to devise a novel molecular classification of PTCLs and to exploit a new therapeutic strategy for this group of aggressive malignancies.
- Published
- 2019
37. Invariant phenotype and molecular association of biallelic TET2 mutant myeloid neoplasia
- Author
-
Babal K. Jha, Bartlomiej P Przychodzen, Yasunobu Nagata, Mikkael A. Sekeres, Cassandra M. Hirsch, Torsten Haferlach, Bhumika J. Patel, Mai Aly, Teodora Kuzmanovic, Jaroslaw P. Maciejewski, Vera Adema, Mohamed E. Abazeed, Abhinav Goyal, Yihong Guan, Valeria Visconte, Louis Williams, Hassan Awada, Mohammad Fahad B Asad, Tomas Radivoyevitch, Aziz Nazha, and Wenyi Shen
- Subjects
Neuroblastoma RAS viral oncogene homolog ,Myeloid ,Chronic myelomonocytic leukemia ,Hematology ,Hyperplasia ,Biology ,medicine.disease ,medicine.disease_cause ,Molecular biology ,Uniparental disomy ,Haematopoiesis ,medicine.anatomical_structure ,Monocytosis ,medicine ,KRAS - Abstract
Somatic TET2 mutations ( TET2 MT ) are frequent in myeloid neoplasia (MN), particularly chronic myelomonocytic leukemia (CMML). TET2 MT includes mostly loss-of-function/hypomorphic hits. Impaired TET2 activity skews differentiation of hematopoietic stem cells toward proliferating myeloid precursors. This study was prompted by the observation of frequent biallelic TET2 gene inactivations ( biTET2 i ) in CMML. We speculated that biTET2 i might be associated with distinct clinicohematological features. We analyzed TET2 MT in 1045 patients with MN. Of 82 biTET2 i cases, 66 were biTET2 MT , 13 were hemizygous TET2 MT , and 3 were homozygous TET2 MT (uniparental disomy); the remaining patients (denoted biTET2 − hereafter) were either monoallelic TET2 MT (n = 96) or wild-type TET2 (n = 823). Truncation mutations were found in 83% of biTET2 i vs 65% of biTET2 − cases ( P = .02). TET2 hits were founder lesions in 72% of biTET2 i vs 38% of biTET2 − cases ( P biTET2 i , significantly concurrent hits included SRSF2 MT (33%; P KRAS / NRAS MT (16%; P = .03) as compared with biTET2 − . When the first TET2 hit was ancestral in biTET2 i , the most common subsequent hits affected a second TET2 MT , followed by SRSF2 MT , ASXL1 MT , RAS MT , and DNMT3A MT . BiTET2 i patients without any monocytosis showed an absence of S RSF2 MT . BiTET2 i patients were older and had monocytosis, CMML, normal karyotypes, and lower-risk disease compared with biTET2 − patients. Hence, while a second TET2 hit occurred frequently, biTET2 i did not portend faster progression but rather determined monocytic differentiation, consistent with its prevalence in CMML. Additionally, biTET2 i showed lower odds of cytopenias and marrow blasts (≥5%) and higher odds of myeloid dysplasia and marrow hypercellularity. Thus, biTET2 i might represent an auxiliary assessment tool in MN.
- Published
- 2019
38. Clonal PIGA mosaicism and dynamics in paroxysmal nocturnal hemoglobinuria
- Author
-
Taha Bat, Cassandra M. Hirsch, Yasunobu Nagata, Marcin W. Wlodarski, Bartlomiej P Przychodzen, Hideki Makishima, Michael J. Clemente, Tomas Radivoyevitch, and Jaroslaw P. Maciejewski
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Adolescent ,Hemoglobinuria, Paroxysmal ,Biology ,Article ,Young Adult ,03 medical and health sciences ,medicine ,Humans ,Child ,Aged ,Aged, 80 and over ,Mosaicism ,Membrane Proteins ,Hematology ,Middle Aged ,medicine.disease ,030104 developmental biology ,Oncology ,Child, Preschool ,Immune System ,Mutation ,Immunology ,Paroxysmal nocturnal hemoglobinuria ,Female - Published
- 2018
39. Distinct gene alterations with a high percentage of myeloperoxidase-positive leukemic blasts in de novo acute myeloid leukemia
- Author
-
Rika Kihara, Tomoki Naoe, Seishi Ogawa, Shigeki Ohtake, Hiroko Tanaka, Rena Kamijo, Norio Asou, Hidehiro Itonaga, Hitoshi Kiyoi, Yuichi Shiraishi, Kenichi Chiba, Tomoko Hata, Yasunobu Nagata, Yasushi Miyazaki, and Satoru Miyano
- Subjects
Adult ,0301 basic medicine ,Cancer Research ,IDH1 ,Adolescent ,animal diseases ,Chimeric gene ,Gene mutation ,medicine.disease_cause ,IDH2 ,Fusion gene ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,CEBPA ,Biomarkers, Tumor ,medicine ,Humans ,RNA, Messenger ,Peroxidase ,Mutation ,Acute myeloid leukemia ,Myeloperoxidase ,Gene Expression Regulation, Leukemic ,business.industry ,Myeloid leukemia ,Hematology ,DNA Methylation ,Middle Aged ,Prognosis ,Molecular biology ,Leukemia, Myeloid, Acute ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,business ,Genes, Neoplasm - Abstract
The myeloperoxidase (MPO)-positivity of blasts in bone marrow smears is an important marker for not only the diagnosis, but also the prognosis of acute myeloid leukemia (AML). To investigate the relationship between genetic alterations and MPO-positivity, we performed targeted sequencing for 51 genes and 10 chimeric gene transcripts in 164 newly diagnosed de novo AML patients; 107 and 57 patients were classified as AML with >50% MPO-positive blasts (MPO-high group) and ?50% MPO-positive blasts, (MPO-low group), respectively. The univariate analysis revealed that RUNX1-RUNX1T1 (P < 0.001), the KIT mutation (P < 0.001), and CEBPA double mutation (P = 0.001) were more likely to be found in the MPO-high group, while the DNMT3A mutation (P = 0.001), FLT3 tyrosine kinase domain mutation (P = 0.004), and TP53 mutation (P = 0.020) were more likely to be present in the MPO-low group. Mutations in genes related to DNA hypermethylation signatures (IDH1, IDH2, TET2, and WT1 genes) were more frequent in the MPO-high group (P = 0.001) when patients with fusion genes of core-binding factors were excluded from the analysis. Our results suggest that MPO-positivity of blasts was related with the distinct gene mutation patterns among de novo AML patients., Leukemia Research, 65, pp.34-41; 2018
- Published
- 2018
40. Prognostic relevance of integrated genetic profiling in adult T-cell leukemia/lymphoma
- Author
-
Tetsuichi Yoshizato, Masao Matsuoka, Kotaro Shide, Wataru Munakata, Makoto Yoshimitsu, Kenichi Yoshida, Hiromichi Suzuki, Kenji Ishitsuka, Hidehiro Itonaga, Hiroko Tanaka, Osamu Nureki, Yasunobu Nagata, Yoko Kubuki, Ryohei Ishii, Tatsuhiro Shibata, Kenichi Chiba, Kazuya Shimoda, Yotaro Ochi, Yasushi Miyazaki, Kosuke Aoki, Kisato Nosaka, Masashi Sanada, Seishi Ogawa, Aiko Sato-Otsubo, Tsuyoshi Nakamaki, Satoru Miyano, Akifumi Takaori-Kondo, Atae Utsunomiya, Keisuke Kataoka, Yusuke Sato, Yusuke Shiozawa, Toshiki Watanabe, Yuichi Shiraishi, Masakatsu Hishizawa, Masako Iwanaga, Ken Ishiyama, Jun-ichirou Yasunaga, Tomonori Hidaka, Shuichi Miyawaki, Takuro Kameda, Yoshitaka Imaizumi, Kensei Tobinai, and Akira Kitanaka
- Subjects
Adult ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lymphoma ,Immunology ,Single-nucleotide polymorphism ,Biology ,Biochemistry ,Gene dosage ,Adult T-cell leukemia/lymphoma ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,CDKN2A ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Epigenetics ,Genotyping ,Lymphoid Neoplasia ,Cell Biology ,Hematology ,Prognosis ,medicine.disease ,Leukemia ,030104 developmental biology ,030220 oncology & carcinogenesis - Abstract
Adult T-cell leukemia/lymphoma (ATL) is a heterogeneous group of peripheral T-cell malignancies characterized by human T-cell leukemia virus type-1 infection, whose genetic profile has recently been fully investigated. However, it is still poorly understood how these alterations affect clinical features and prognosis. We investigated the effects of genetic alterations commonly found in ATL on disease phenotypes and clinical outcomes, based on genotyping data obtained from 414 and 463 ATL patients using targeted-capture sequencing and single nucleotide polymorphism array karyotyping, respectively. Aggressive (acute/lymphoma) subtypes were associated with an increased burden of genetic and epigenetic alterations, higher frequencies of TP53 and IRF4 mutations, and many copy number alterations (CNAs), including PD-L1 amplifications and CDKN2A deletions, compared with indolent (chronic/smoldering) subtypes. By contrast, STAT3 mutations were more characteristic of indolent ATL. Higher numbers of somatic mutations and CNAs significantly correlated with worse survival. In a multivariate analysis incorporating both clinical factors and genetic alterations, the Japan Clinical Oncology Group prognostic index high-risk, older age, PRKCB mutations, and PD-L1 amplifications were independent poor prognostic factors in aggressive ATL. In indolent ATL, IRF4 mutations, PD-L1 amplifications, and CDKN2A deletions were significantly associated with shorter survival, although the chronic subtype with unfavorable clinical factors was only marginally significant. Thus, somatic alterations characterizing aggressive diseases predict worse prognosis in indolent ATL, among which PD-L1 amplifications are a strong genetic predictor in both aggressive and indolent ATL. ATL subtypes are further classified into molecularly distinct subsets with different prognosis. Genetic profiling might contribute to improved prognostication and management of ATL patients.
- Published
- 2018
41. Epigenetic Enzyme Mutations in Myeloid Malignancies Are Selected By Chromatin-Remodeling Requirements That Vary By Lineage- and Maturation-Stage
- Author
-
Sunisa Kongkiatkamon, Xiaorong Gu, Kwok Peng Ng, Simona Pagliuca, Vera Adema, Wencke Walter, Yasunobu Nagata, Cassandra M Kerr, Manja Meggendorfer, Hassan Awada, Stephan Hutter, Carmelo Gurnari, Claudia Haferlach, Babal K. Jha, Valeria Visconte, Torsten Haferlach, Jaroslaw P. Maciejewski, and Yogenthiran Saunthararajah
- Subjects
chemistry.chemical_classification ,Myeloid ,Lineage (genetic) ,Immunology ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Chromatin remodeling ,Cell biology ,medicine.anatomical_structure ,Enzyme ,chemistry ,hemic and lymphatic diseases ,medicine ,Epigenetics ,Stage (cooking) - Abstract
Introduction/Methods: Enzymes that modify histone H3 at lysine 27 (H3K27) to thereby regulate gene transcription (epigenetic enzymes) are recurrently inactivated by deletion and/or mutation in myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPN) and acute myeloid leukemias (AML). Frustrating understanding of mechanisms is that writers and erasers of the same modification, e.g., methyltransferases that create, and demethylases that remove, H3K27 trimethylation (H3K27me3), a repression ('off') mark, are recurrently inactivated. Moreover, acetyltransferases that write H3K27 acetylation (H3K27ac), an 'on' mark mutually exclusive with H3K27me3, are also recurrently inactivated. One clue to underlying mechanisms is that MDS/MPN/AML present with diverse lineage and maturation phenotypes - perhaps these emerge from, or select for, the diverse epigenetic enzyme mutations. We therefore identified genes upregulated with specific myeloid lineage-commitment, maturation and function fates (~500 genes each) and then examined distributions of H3K27me3 and H3K27ac at these gene-loci in: (i) embryonic stem cells (ESC); (ii) hematopoietic stem and progenitor cells (HSPC); (iii) mature myeloid cells (monocyte [mono], pro-erythroblast, megakaryocyte [MK]); and (iv) AML cells. Results: Terminal-myeloid programs underwent substantial remodeling to gain H3K27ac 'on' mark from ESC/HSPC to mature myeloid (Fig.1A). Providing a mechanism for this, the H3K27 acetyltransferases EP300 and CREBBP were recruited into the RUNX1/SPI1 myeloid-lineage master transcription factor (MTF) hub by cooperation between their transcription activating domains. Mutated/translocated RUNX1, or mutated-NPM1 that cytoplasmically dislocated SPI1, disrupted this cooperation, and reverted hub content to default recruitment of histone deacetylases (HDAC) instead. Demonstrating cause-effect, inhibiting these HDAC renewed AML cell maturation to terminal lineage-fates. Meanwhile, MYC-target (proliferation) genes have high baseline H3K27ac in ESC and HSPC and do not require major remodeling during ontogeny (Fig.1A). An H3K27ac remodeling requirement for lineage-maturation but not proliferation/housekeeping explains selection pressure for inactivating mutations in EP300 or CREBBP, that we found in ~1.2% of MDS/MPN/AML in our (n=690) and other series. Consistent with pan-lineage-maturation needs for H3K27ac, the mutations were found in all lineage sub-types. H3K27me3 'off' mark was mostly erased from myeloid programs in HSPC, but was greater at MK vs erythroid, and also at mono vs granulocyte genes (Fig.1B). This implied more need for H327me3 demethylase (KDM6A/UTX) for HSPC commitment into MK vs erythroid, or mono vs granulocyte, lineages. Accordingly, KDM6A was most upregulated in MK and mono-lineage cells (Fig.1C), and myeloid-conditional Kdm6a knockout decreased platelets and increased red cells in the spleen - reported by others: https://doi.org/10.1182/blood.V128.22.1467.1467. RUNX1-ETO has been shown to specifically impede granulocytic but not mono differentiation - https://www.nature.com/articles/2403396 and KDM6A inactivating mutations were significantly more likely to occur secondary to RUNX1-ETO (4-9% BEAT and AMLSG case series) vs other cytogenetics (0.005%). Selection for KDM6A secondary mutations could thus be to channel myeloid precursors toward lineages most efficiently impeded by primary mutations. Although H3K27me3 was substantially erased at all myeloid-commitment and terminal programs (except MK) in HSPC vs ESC, subsequent lineage-commitment and maturation entailed rewriting H3K27me3 at preceding HSPC and alternate lineage-fate programs, including at MTF genes for alternate fates (Fig.1B, D). Primary MDS/MPN/AML and AML cell lines with inactivating mutations/deletions in H3K27 methyltransferase EZH2 thus displayed aberrant co-expression of lineage MTFs and gene expression programs of normally mutually exclusive lineages (Fig.1E-G). Conclusion. Epigenetic remodeling requirements vary by myeloid lineage and maturation stage. Thus, epigenetic enzyme mutations are selected by, and cause, lineage-context of transformation. This knowledge can guide choice of specific epigenetic enzyme inhibitors to remedy the lineage-maturation defects that drive and define myeloid malignancies. Figure 1 Figure 1. Disclosures Haferlach: MLL Munich Leukemia Laboratory: Other: Part ownership. Haferlach: MLL Munich Leukemia Laboratory: Other: Part ownership. Maciejewski: Novartis: Consultancy; Bristol Myers Squibb/Celgene: Consultancy; Regeneron: Consultancy; Alexion: Consultancy. Saunthararajah: EpiDestiny: Consultancy, Current holder of individual stocks in a privately-held company, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties.
- Published
- 2021
42. EPOR/JAK/STAT Signaling Pathway As Therapeutic Target of Acute Erythroid Leukemia
- Author
-
Kensuke Usuki, Toshiyuki Kitano, Hiroko Tanaka, Akifumi Takaori-Kondo, Nobuhiro Hiramoto, Shuichi Miyawaki, Arnold Ganser, Yuichi Shiraishi, Seishi Ogawa, Hisashi Tsurumi, Jaroslaw P. Maciejewski, Kenichi Chiba, Satoru Miyano, Yasunobu Nagata, Yasushi Miyazaki, Ming-Chung Kuo, June Takeda, Yasuhito Nannya, Kazuma Ohyashiki, Akira Hangaishi, Shigeru Chiba, Michael Heuser, Akinori Yoda, Ken Ishiyama, Lee-Yung Shih, Cassandra M Kerr, Ryunosuke Saiki, Felicitas Thol, Kenichi Yoshida, Hideki Makishima, Daisuke Morishita, Hideyuki Nakazawa, Masahiro Nakagawa, Takayuki Ishikawa, and Nobuo Sezaki
- Subjects
business.industry ,Immunology ,Cancer research ,Acute erythroid leukemia ,JAK-STAT signaling pathway ,Medicine ,Cell Biology ,Hematology ,business ,medicine.disease ,Biochemistry ,Erythropoietin receptor - Abstract
Background: Acute erythroid leukemia (AEL) is a rare subtype of AML characterized by erythroid predominant proliferation and classified into two subtypes with pure erythroid (PEL) and myeloid/erythroid (MEL) phenotypes. Although several reports described gene mutations in AEL, genotype phenotype correlations have not fully been elucidated with little knowledge about feasible molecular targets for therapy. Methods: To understand the mechanism of the erythroid dominant phenotype of AEL and identify potential therapeutic targets for AEL, we analyzed a total of 121 adult AEL cases with the median age of 60 (23-87), using whole genome/exome sequencing of 35 cases, followed by targeted-capture sequencing of 387 genes together with 1,279 SNP loci for copy number measurements in all cases. Among these, 21 were also analyzed by RNA sequencing. Genetic profiles of these AEL cases were compared to those of 409 cases with non-erythroid AML (non-AEL) including 195 cases from The Cancer Genome Atlas. Six patient-derived xenografts (PDX) were established from AEL with JAK2 and/or EPOR focal gain/amplification/mutation. PDX cells were inoculated into immune-deficient mice and tested for their response to JAK1/2 inhibitor. Results: According to unique genetic alterations, AEL was classified into 4 genomic groups (A-D). Characterized by TP53 mutations and complex karyotype, Group A was the most common subtype (48/121; 40%) and showed very poor prognosis. Remarkably, almost all the PEL cases (12/13; 92%) were categorized into Group A. Conspicuously, 75% of PEL cases with TP53 mutation had focal gain/amplifications/mutations of JAK2 (5/12; 42%), EPOR (7/12; 58%), and ERG/ETS2 (1/12; 8%) loci on chromosomes 9p, 19q, and 21q, respectively, while 34% of MEL cases with TP53 mutation had focal gain/amplifications/mutations of JAK2 (2/29; 7%), EPOR (7/29;24%), and ERG/ETS2 (7/29;24%) loci, frequently in combination. Group B was characterized by frequent NPM1 mutations, in contrast to the frequent co-mutation of FLT3 in the corresponding subgroup of NPM1-mutated cases in non-AEL, whereas NPM1-mutated patents in this group lacked FLT3 mutations but had frequent PTPN11 mutations (8/16; 50%), which were much less common in non-AEL (15/101; 15%). All cases in Group C (n=22, 18%), another prevalent form of AEL, had STAG2 mutations and classified in MEL. Prominently, 68% (17/25) of STAG2-mutated AEL cases had KMT2A-PTD, which was rarely found in non-AEL cases. The remaining cases were categorized into Group D, which was enriched for mutations in ASXL1, BCOR, PHF6, RUNX1 and TET2. We also identified recurrent loss-of-function USP9X mutations in this group, which were previously reported in ALL with an upregulated JAK-STAT pathway. In RNA sequencing analysis, AEL cases exhibited gene expression profiles implicated in an upregulated STAT5 signaling pathway, which was seen not only in those cases with JAK2 or EPOR focal gain/amplification/mutation, but also in AEL without these amplifications, suggesting that aberrantly upregulated STAT5 activation might represent a common molecular signature of AEL. Survival analysis revealed that TP53 mutation is a poor prognostic factor in AEL and non-AEL and no statistically significant difference between AEL and non-AEL with TP53 mutation. Intriguingly, 19p gains/amplifications were associated with a significantly poor prognostic prognosis in TP53-mutated AEL cases. Based on this finding, we evaluated the effect of a JAK inhibitor, ruxolitinib, on 6 PDX models established from AEL having TP53 mutations and JAK2 and EPOR mutation/amplification. Of interest , ruxolitinib significantly suppressed cell growth and prolonged overall survival in mice engrafted with 4 PDX models with STAT5 downregulation, although the other 2 models were resistant to JAK2 inhibition with persistent STAT5 activation. Conclusion: AEL is a heterogeneous group of AML, of which PEL is characterized by frequent amplifications/mutations in JAK2 and/or EPOR. Frequent involvement of EPOR/JAK/STAT pathway is a common feature of AEL, in which a therapeutic role of JAK inhibition was suggested. Disclosures Nakagawa: Sumitomo Dainippon Pharma Oncology, Inc.: Research Funding. Yoda: Chordia Therapeutics Inc.: Research Funding. Morishita: Chordia Therapeutics Inc.: Current Employment, Current equity holder in publicly-traded company. Miyazaki: Sumitomo-Dainippon: Honoraria, Research Funding; Astellas: Honoraria; Chugai: Honoraria; Abbvie: Honoraria; Novartis: Honoraria; Nippon-Shinyaku: Honoraria; Bristol-Myers Squibb: Honoraria; Takeda: Honoraria; Daiichi-Sankyo: Honoraria; Kyowa-Kirin: Honoraria; Eisai: Honoraria; Janssen: Honoraria; Pfizer: Honoraria; Sanofi: Honoraria. Usuki: Alexion: Speakers Bureau; Eisai: Speakers Bureau; MSD: Speakers Bureau; PharmaEssentia: Speakers Bureau; Yakult: Speakers Bureau; Mundipharma: Research Funding; Astellas-Amgen-Biopharma: Research Funding; Nippon Boehringer Ingelheim: Research Funding; Takeda: Research Funding, Speakers Bureau; Celgene: Research Funding, Speakers Bureau; Janssen: Research Funding; Ono: Research Funding, Speakers Bureau; Otsuka: Research Funding, Speakers Bureau; Sumitomo Dainippon: Research Funding; Daiichi Sankyo: Research Funding, Speakers Bureau; Symbio: Research Funding, Speakers Bureau; Gilead: Research Funding; Abbvie: Research Funding; Nippon shinyaku: Research Funding, Speakers Bureau; Novartis: Research Funding, Speakers Bureau; Pfizer: Research Funding; Kyowa Kirin: Research Funding, Speakers Bureau; Brisol-Myers Squibb: Research Funding, Speakers Bureau; Astellas: Research Funding, Speakers Bureau. Maciejewski: Bristol Myers Squibb/Celgene: Consultancy; Regeneron: Consultancy; Novartis: Consultancy; Alexion: Consultancy. Ohyashiki: Novartis Pharma: Other: chief clinical trial; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees. Ganser: Celgene: Honoraria; Novartis: Honoraria; Jazz Pharmaceuticals: Honoraria. Heuser: Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer Pharma AG: Research Funding; Karyopharm: Research Funding; Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees, Research Funding; BergenBio: Research Funding; Janssen: Honoraria; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Astellas: Research Funding; AbbVie: Membership on an entity's Board of Directors or advisory committees, Research Funding; Tolremo: Membership on an entity's Board of Directors or advisory committees; Jazz: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS/Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Membership on an entity's Board of Directors or advisory committees, Research Funding. Thol: Astellas: Honoraria; Abbvie: Honoraria; Novartis: Honoraria; Jazz: Honoraria; BMS/Celgene: Honoraria, Research Funding; Pfizer: Honoraria. Shih: PharmaEssentia Co: Consultancy, Membership on an entity's Board of Directors or advisory committees; Celgene Ltd: Research Funding; Ltd: Research Funding; Novartis: Research Funding. Takaori-Kondo: Celgene: Research Funding; Bristol-Myers K.K.: Honoraria; ONO PHARMACEUTICAL CO., LTD.: Research Funding. Ogawa: Otsuka Pharmaceutical Co., Ltd.: Research Funding; Eisai Co., Ltd.: Research Funding; Kan Research Laboratory, Inc.: Consultancy, Research Funding; Dainippon-Sumitomo Pharmaceutical, Inc.: Research Funding; ChordiaTherapeutics, Inc.: Consultancy, Research Funding; Ashahi Genomics: Current holder of individual stocks in a privately-held company.
- Published
- 2021
43. Der(1;7)(q10;p10) Presents with a Unique Genetic Profile and Frequent ETNK1 Mutations in Myeloid Neoplasms
- Author
-
Claudia Haferlach, Yasunobu Nagata, June Takeda, Yoshiko Atsuta, Senji Kasahara, Niroshan Nadarajah, Hiroshi Handa, Masashi Sanada, Wolfgang Kern, Satoru Miyano, Constance Baer, Kenichi Yoshida, Yuichi Shiraishi, Yotaro Ochi, Torsten Haferlach, Shigeru Chiba, Kazuma Ohyashiki, Yasuhito Nannya, Rurika Okuda, Hideki Makishima, Ayana Kon, Seishi Ogawa, Maria Creignou, Tetsuichi Yoshizato, and Eva Hellstrom Lindberg
- Subjects
Genetics ,Myeloid ,medicine.anatomical_structure ,Immunology ,medicine ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Genetic profile - Abstract
Background Der(1;7)(q10;p10) (der(1;7) is an unbalanced translocation recurrently found in myeloid neoplasms, particularly in myelodysplastic syndromes (MDS) and related disorders. Caused by a recombination between two homologous alphoid sequencing D1Z7 and D7Z1 on chromosomes 1 and 7, respectively, it results in monosomy 7q and trisomy 1q, which is implicated in the pathogenesis of der(1;7)-positive myeloid neoplasms. Previous studies reported frequent co-occurrence of +8 and del(20q), as well as RUNX1 mutations, the genetic and clinical characteristics of this abnormality has not fully been elucidated. Methods In this study, we enrolled a total of 153 cases myeloid neoplasms positive for der(1;7) from Japanese and German cohorts, in which co-occurring genetic lesions were analyzed using whole exome and/or targeted-capture sequencing. An additional 3,223 MDS and related neoplasm cases were also analyzed using targeted-capture sequencing to identify der(1;7)-specific genomic features. Results Ethnicity was evaluated comparing the frequency of der(1;7) in 944 German MDS cases and 763 Japanese MDS cases. Der(1;7) cases were observed at a higher frequency in Japanese MDS cohort compared to German MDS cohort (73/763 cases versus 4/944 cases, p < 0.00001). Der(1;7) cases showed a strong male predominance (86.3%) (p Survival analysis was conducted to elucidate the difference in survival in der(1;7) cases (n=65) versus myeloid neoplasm cases (n=2066). Der(1;7)-harboring myeloid neoplasm cases had a median overall survival of 6.8 months (95% CI, 3.5 to 11.9) and non-der(1;7) harboring myeloid neoplasm cases were 11.8 months (95% CI, 10.5 to 12.6). Thus, der(1;7)-harboring myeloid neoplasm cases had poorer prognosis (p Conclusion In conclusion, der(1;7) is an unbalanced translocation that occurs predominantly in males and is seen more frequently in Japanese than Caucasian populations. Der(1;7) cases present with a mutational profile that is distinct from other myeloid neoplasm cases such as those with amp(1q) and monosomy7/del(7q), showing frequency of ETNK1 mutations. Disclosures Nannya: Otsuka Pharmaceutical Co., Ltd.: Consultancy, Speakers Bureau; Astellas: Speakers Bureau. Kern: MLL Munich Leukemia Laboratory: Other: Part ownership. Haferlach: MLL Munich Leukemia Laboratory: Other: Part ownership. Atsuta: Astellas Pharma Inc.: Speakers Bureau; Mochida Pharmaceutical Co., Ltd.: Speakers Bureau; AbbVie GK: Speakers Bureau; Kyowa Kirin Co., Ltd: Honoraria; Meiji Seika Pharma Co, Ltd.: Honoraria. Handa: Ono: Honoraria; BMS: Honoraria; Janssen: Honoraria; Daiichi Sankyo: Research Funding; Celgene: Honoraria, Research Funding; Chugai: Research Funding; Kyowa Kirin: Research Funding; Takeda: Honoraria, Research Funding; Sanofi: Honoraria, Research Funding; Abbvie: Honoraria; MSD: Research Funding; Shionogi: Research Funding. Ohyashiki: Novartis Pharma: Other: chief clinical trial; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees. Haferlach: MLL Munich Leukemia Laboratory: Other: Part ownership. Ogawa: Otsuka Pharmaceutical Co., Ltd.: Research Funding; Eisai Co., Ltd.: Research Funding; Kan Research Laboratory, Inc.: Consultancy, Research Funding; Dainippon-Sumitomo Pharmaceutical, Inc.: Research Funding; ChordiaTherapeutics, Inc.: Consultancy, Research Funding; Ashahi Genomics: Current holder of individual stocks in a privately-held company.
- Published
- 2021
44. Germline MPO Variants Predispose to Myeloid Neoplasia: Potential Mechanisms Suggested By In Vivo and in Vitro Studies
- Author
-
Metis Hasipek, Yasunobu Nagata, Sunisa Kongkiatkamon, Babal K. Jha, Milo Co, Daniel J. Lindner, Yihong Guan, Yvonne Parker, Vera Adema, Carmelo Gurnari, Stephan Hutter, Wencke Walter, Hassan Awada, Tatiana Dombrovski, Valeria Visconte, Cassandra M Kerr, Jaroslaw P. Maciejewski, Manja Meggendorfer, Simona Pagliuca, Torsten Haferlach, Thomas LaFramboise, and Laila Terkawi
- Subjects
Myeloid neoplasia ,In vivo ,Immunology ,Cancer research ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Germline ,In vitro - Abstract
In recent decades, there has been a substantial increase in recognition of germline (GL) predisposition to MN. Some congenital blood disorders predispose to subsequent myeloid neoplasia (MN). Severe congenital neutropenia (SCN) and Fanconi anemia serving as prominent examples. There is emerging evidence that heterozygous variants of disease recessive conditions may constitute risk alleles for late developing MN. However, their low penetrance may preclude identification of such alterations. Our previous analysisof allelic burden among selected combined potential GL predisposition genes to MN identified that the myeloperoxidase (MPO) gene (17q22-23) carried the highest risk of pathogenic alterations (Li, S.T, Leukemia, 2020). GL MPO mutations cause MPO deficiency syndrome, one of the most common inherited disorder associated with phagocyte defects and variable clinical penetrance. Despite the high prevalence, inherited MPO deficiency patients are asymptomatic and may remain unrecognized. We investigated 3280 MN and bone marrow failure syndromes (BMF) pts including 1138 MDS, 260 MDS/MPN, 1661 AML and 221 AA for presence of MPO mutations. In total, 38 different germline MPO mutations were identified in 143 cases. With a stringent bioanalytic pipeline, 28 pathogenic/likely pathogenic variants from 100 MN patients were included in the study. Germline MPO variants were significantly enriched in MN compared to control population (294 vs. 125 per 10 4 individuals; PC) followed by R569W (13/100), M519fs* (13/100) and Y173C (6/100) with none being biallelic. While no differences were found in distribution of -7/7q, del5q, or del20q, MPO variants carriers harbored less tri-8 compared to wild type counterparts (2% vs. 12%; P=.008). Only FLT3 and NRAS were significantly associated with MPO mutations. We then investigated the effects of MPO deficiency on hematopoietic function in murine model using competitive repopulation assays, whereby the difference in CD45.1/CD45.2 isotypes transplanted in to ROSA26 (tdTomato-EGFP) recipients assayed by flow cytometry allowed distinction of the 2 grafts. Mpo-/- cells gained over time proliferative advantage over normal murine bone marrow cells. Reverse combinations (graft mix vs recipient) also replicated this result. This effect was not due to increased HSC content in Mpo-/- marrow as there were no significant differences in LSK, CMP and MEPs cells. We then investigated the clonogenic consequences of Mpo-/- cells in the setting of H 2O 2 induced oxidative stress, after exposing to hydrogen peroxide. Mpo-/- cells increased clonogenic potential after second serially replating. This prompted us to investigate an MPO inhibitor, AZD5904 in human leukemia cells with different MPO expression (HL-60, high; K562, low). High MPO expressors retained higher cell viability following H 2O 2 and MPOi addition compared to those without MPOi (85±10 vs.59±8, P In conclusion, for the first time we demonstrated that germline MPO variants constitute risk alleles for MN evolution and replicated the potential replicative advantage of MPO deficient cells in murine model and potential mechanisms of the MPO deficiency in vitro including activation of non-homologous DNA repair response and error-prone DNA repair favoring replication. Repeated cycles of stress hematopoiesis e.g., due to increased infection rate may provide conditions in which MPO variants contribute to the risk of MN. Disclosures Haferlach: MLL Munich Leukemia Laboratory: Other: Part ownership. Maciejewski: Regeneron: Consultancy; Alexion: Consultancy; Bristol Myers Squibb/Celgene: Consultancy; Novartis: Consultancy.
- Published
- 2021
45. Origins of myelodysplastic syndromes after aplastic anemia
- Author
-
Yasunobu Nagata, Teodora Kuzmanovic, Seishi Ogawa, Bartlomiej P Przychodzen, Tetsuichi Yoshizato, Reda Z. Mahfouz, Eiju Negoro, Aziz Nazha, Michael J. Clemente, Wenyi Shen, Jaroslaw P. Maciejewski, Cassandra M. Hirsch, Hideki Makishima, Naoko Hosono, and Mikkael A. Sekeres
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Anemia ,Immunology ,Hemoglobinuria, Paroxysmal ,Biochemistry ,Clonal Evolution ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Humans ,Aplastic anemia ,Letter to Blood ,Extramural ,business.industry ,Myelodysplastic syndromes ,Disease progression ,Secondary Myelodysplastic Syndrome ,Anemia, Aplastic ,Cell Biology ,Hematology ,medicine.disease ,Dermatology ,030104 developmental biology ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Mutation ,Disease Progression ,Paroxysmal nocturnal hemoglobinuria ,Hemoglobinuria ,business - Abstract
To the editor: The course of aplastic anemia (AA) is often complicated by the development of clonal disorders such as paroxysmal nocturnal hemoglobinuria (PNH) and secondary myelodysplastic syndromes (sMDS).[1][1][⇓][2][⇓][3][⇓][4]-[5][5] Identification of patients at risk for development of
- Published
- 2017
46. Distinct clinical and biological implications of various DNMT3A mutations in myeloid neoplasms
- Author
-
Cassandra M. Hirsch, Valeria Visconte, Vera Adema, Suresh Kumar Balasubramanian, Sudipto Mukherjee, Mikkael A. Sekeres, Bartlomiej P Przychodzen, Tomas Radivoyevitch, Taha Bat, Mai Aly, Yasunobu Nagata, Jaroslaw P. Maciejewski, Teodora Kuzmanovic, and Aziz Nazha
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Myeloid ,Computational biology ,Biology ,Article ,DNA Methyltransferase 3A ,03 medical and health sciences ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Humans ,DNA (Cytosine-5-)-Methyltransferases ,Aged ,Hematology ,Prognosis ,Leukemia, Myeloid, Acute ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,embryonic structures ,Female - Abstract
Distinct clinical and biological implications of various DNMT3A mutations in myeloid neoplasms
- Published
- 2017
47. Molecular features of early onset adult myelodysplastic syndrome
- Author
-
Thomas LaFramboise, Hetty E. Carraway, Tomas Radivoyevitch, Mikkael A. Sekeres, Cassandra M. Hirsch, Bhumika J. Patel, Hideki Makishima, Yasunobu Nagata, Swapna Thota, Jaroslaw P. Maciejewski, Aziz Nazha, Michael J. Clemente, and Bartlomiej P Przychodzen
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Myeloid ,DNA Mutational Analysis ,Disease ,Biology ,Article ,DNA Methyltransferase 3A ,Dioxygenases ,Pathogenesis ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Proto-Oncogene Proteins ,Internal medicine ,medicine ,Humans ,Secondary Acute Myeloid Leukemia ,DNA (Cytosine-5-)-Methyltransferases ,Adult Myelodysplastic Syndrome ,Age of Onset ,Young adult ,10. No inequality ,Aged ,Aged, 80 and over ,Serine-Arginine Splicing Factors ,Myelodysplastic syndromes ,Age Factors ,High-Throughput Nucleotide Sequencing ,Hematology ,Middle Aged ,medicine.disease ,3. Good health ,DNA-Binding Proteins ,030104 developmental biology ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,030220 oncology & carcinogenesis ,Mutation ,Immunology ,Female ,Age of onset - Abstract
Myelodysplastic syndromes are typically diseases of older adults. Patients in whom the onset is early may have distinct molecular and clinical features or reflect a demographic continuum. The identification of differences between “early onset” patients and those diagnosed at a traditional age has the potential to advance understanding of the pathogenesis of myelodysplasia and may lead to formation of distinct morphological subcategories. We studied a cohort of 634 patients with various subcategories of myelodysplastic syndrome and secondary acute myeloid leukemia, stratifying them based on age at presentation and clinical parameters. We then characterized molecular abnormalities detected by next-generation deep sequencing of 60 genes that are commonly mutated in myeloid malignancies. The number of mutations increased linearly with age and on average, patients >50 years of age had more mutations. TET2, SRSF2, and DNMT3A were more commonly mutated in patients >50 years old compared to patients ≤50 years old. In general, patients >50 years of age also had more mutations in spliceosomal, epigenetic modifier, and RAS gene families. Although there are age-related differences in molecular features among patients with myelodysplasia, most notably in the incidence of SRSF2 mutations, our results suggest that patients ≤50 years old belong to a disease continuum with a distinct pattern of early onset ancestral events.
- Published
- 2017
48. Genotype-Phenotype Relationships and Therapeutic Targets in Acute Erythroid Leukemia
- Author
-
Ming-Chung Kuo, Kenichi Yoshida, Akinori Yoda, Ayana Kon, Seishi Ogawa, Cassandra M Kerr, Kenichi Chiba, Yasunobu Nagata, Yasushi Miyazaki, Ken Ishiyama, Felicitas Thol, Hiroko Tanaka, Arnold Ganser, Ryunosuke Saiki, Toshiyuki Kitano, Nobuhiro Hiramoto, Shuichi Miyawaki, Michael Heuser, Yotaro Ochi, Hideyuki Nakazawa, Satoru Miyano, Masahiro Nakagawa, Yasuhito Nannya, Hisashi Tsurumi, Yuichi Shiraishi, Keisuke Kataoka, Yusuke Shiozawa, Akira Hangaishi, Hideki Makishima, Tetsuichi Yoshizato, Lee-Yung Shih, Takayuki Ishikawa, Jaroslaw P. Maciejewski, Kensuke Usuki, June Takeda, Akifumi Takaori-Kondo, and Shigeru Chiba
- Subjects
Oncology ,medicine.medical_specialty ,Poor prognosis ,Ruxolitinib ,business.industry ,Immunology ,Acute erythroid leukemia ,Cell Biology ,Hematology ,Gene mutation ,Tp53 mutation ,medicine.disease ,Biochemistry ,Genotype phenotype ,Internal medicine ,CEBPA ,Medicine ,business ,Genotype-Phenotype Correlations ,health care economics and organizations ,medicine.drug - Abstract
Background: Acute erythroid leukemia (AEL) is a rare subtype of AML characterized by erythroid predominant proliferation and classified into two subtypes with pure erythroid (PEL) and myeloid/erythroid (MEL) phenotypes. Although gene mutations in AEL have been described in several reports, genotype phenotype correlations are not fully understood with little knowledge about the feasible molecular targets for therapy. Methods: To understand the mechanism of the erythroid dominant phenotype of AEL and identify potential therapeutic targets for AEL, we analyzed a total of 105 AEL cases with the median age of 60 (23-86), using targeted-capture sequencing of commonly mutated genes in myeloid neoplasms, together with 1,279 SNPs for copy number measurements. Among these 105 cases, 13 were also analyzed by RNA sequencing. Genetic profiles of these 105 AEL cases were compared to those of 775 cases with non-erythroid AML (NEL) including 561 cases from The Cancer Genome Atlas and Beat AML study. An immature erythroid cell line (TF1) and three patient-derived xenografts (PDX) established from AEL with JAK2 and/or EPOR amplification. Cell line and samples from patients were inoculated into immune-deficient mice and tested for their response to JAK1/2 inhibitor. Results: According to unique genetic alterations, AEL was classified into 4 subgroups (A-D). Characterized by TP53 mutations and complex karyotype, Group A was the most common subtype and showed very poor prognosis. Remarkably, all PEL cases were categorized into Group A. Conspicuously, 80% of PEL cases had amplifications of JAK2 (6/10; 60%), EPOR (7/10;70%), and ERG (6/10;60%) loci on chromosomes 9p, 19q, and 21q, respectively, frequently in combination, although they were rarely seen in NEL cases. All cases in Group B (n=19, 18%), another prevalent form of AEL, had STAG2 mutations and classified in MEL. To further characterize this subgroup, we compared genetic profiles of STAG2-mutated AEL and NEL. Prominently, 70% (14/20) of STAG2-mutated cases in AEL had KMT2A-PTD, whereas it was found only in 8.8% (3/34) of NEL. CEBPA mutations were also more common in AEL (6/21; 29%) than NEL (4/34; 12%). While Group C was characterized by frequent NPM1 mutations, in contrast to the frequent co-mutation of FLT3 in the corresponding subgroup of NPM1-mutated cases in NEL, NPM1-mutated patents in this subgroup lacked FLT3 mutations but had frequent PTPN11 mutations (8/16; 50%), which were much less common in NEL (25/209; 12%). The remaining cases were categorized into Group D, which was enriched for mutations in ASXL1, BCOR, PHF6, U2AF1 and KMT2C. Recurrent loss-of-function mutations in USP9X were unique to this subtype, although USP9X mutations have been reported in ALL with upregulation of JAK-STAT pathway. In RNA sequencing analysis, AEL cases exhibited gene expression profiles implicated in an upregulated STAT5 signaling pathway, which was seen not only those cases with JAK2 or EPOR amplification, but also those without, suggesting that aberrantly upregulated STAT5 activation might represent a common defect in AEL. Based on this finding, we evaluated the effect of a JAK inhibitior, ruxolitinib, on an AEL-derived cell line and three PDX models established from AEL having TP53 mutations and JAK2 and EPOR mutation/amplification. Of interest, ruxolitinib significantly suppressed cell growth and prolonged overall survival in mice engrafted with TF1 and 2 PDX models with STAT5 downregulation, although the other model was resistant to JAK2 inhibition with persistent STAT5 activation. Conclusion: AEL is a heterogeneous group of AML, of which PEL is characterized by frequent amplifications/mutations in JAK2, EPOR and/or ERG. Frequent involvement of EPOR/JAK/STAT pathway is a common feature of AEL, in which a role of JAK inhibition was suggested. Disclosures Yoda: Chordia Therapeutics Inc.: Research Funding. Shih:Novartis: Research Funding; Celgene: Research Funding; PharmaEssentia: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Consultancy, Membership on an entity's Board of Directors or advisory committees. Ishiyama:Alexion: Research Funding; Novartis: Honoraria. Miyazaki:Astellas Pharma Inc.: Honoraria; Sumitomo Dainippon Pharma Co., Ltd.: Honoraria; NIPPON SHINYAKU CO.,LTD.: Honoraria; Celgene: Honoraria; Otsuka Pharmaceutical: Honoraria; Chugai Pharmaceutical Co., Ltd.: Honoraria; Novartis Pharma KK: Honoraria; Kyowa Kirin Co., Ltd.: Honoraria. Nakagawa:Sumitomo Dainippon Pharma Co., Ltd.: Research Funding. Takaori-Kondo:Celgene: Honoraria, Research Funding; Ono Pharmaceutical: Research Funding; Thyas Co. Ltd.: Research Funding; Takeda: Research Funding; CHUGAI: Research Funding; OHARA Pharmaceutical: Research Funding; Sanofi: Research Funding; Novartis Pharma: Honoraria; Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Research Funding; Otsuka Pharmaceutical: Research Funding; Eisai: Research Funding; Astellas Pharma: Honoraria, Research Funding; Kyowa Kirin: Honoraria, Research Funding; Nippon Shinyaku: Research Funding; MSD: Honoraria. Kataoka:Asahi Genomics: Current equity holder in private company; Otsuka Pharmaceutical: Research Funding; Takeda Pharmaceutical Company: Research Funding; CHUGAI PHARMACEUTICAL CO., LTD.: Research Funding. Usuki:Alexion: Research Funding, Speakers Bureau; Apellis: Research Funding; Novartis: Research Funding, Speakers Bureau; Chugai: Research Funding. Maciejewski:Novartis, Roche: Consultancy, Honoraria; Alexion, BMS: Speakers Bureau. Ganser:Novartis: Consultancy; Celgene: Consultancy. Thol:Daiichi Sankyo: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees; Astellas: Membership on an entity's Board of Directors or advisory committees; Abbvie: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees. Ogawa:Sumitomo Dainippon Pharma Co., Ltd.: Research Funding; Otsuka Pharmaceutical Co., Ltd.: Research Funding; Asahi Genomics Co., Ltd.: Current equity holder in private company; Eisai Co., Ltd.: Research Funding; Chordia Therapeutics, Inc.: Membership on an entity's Board of Directors or advisory committees, Research Funding; KAN Research Institute, Inc.: Membership on an entity's Board of Directors or advisory committees, Research Funding. OffLabel Disclosure: Ruxolitinib is used for drug efficacy test using patient-derived xenografts established from acute erythroid leukemia.
- Published
- 2020
49. Rare Germline Alterations of Myeloperoxidase Predispose to Myeloid Neoplasms and Are Associated with Increased Circulating Burden of Microbial DNA
- Author
-
Hassan Awada, Laila Terkawi, Carmelo Gurnari, Cassandra M Kerr, Jaroslaw P. Maciejewski, Thomas LaFramboise, Valeria Visconte, Wencke Walter, Claudia Haferlach, Torsten Haferlach, Samuel Li, Simona Pagliuca, Stephan Hutter, Tatiana Dombrovski, Bartlomiej P Przychodzen, Manja Meggendorfer, Sunisa Kongkiatkamon, Vera Adema, and Yasunobu Nagata
- Subjects
Myeloid ,medicine.anatomical_structure ,biology ,Microbial DNA ,Myeloperoxidase ,Immunology ,biology.protein ,Cancer research ,medicine ,Cell Biology ,Hematology ,Biochemistry ,Germline - Abstract
Germline alterations causing inherited myeloid neoplasia (MN) have become prominent in the 2016 WHO classification. Increasing evidence reveals that many patients with MN (MDS and AML) may indeed harbor unsuspected predisposing or direct driver germline alterations. Penetrance of such alterations may be variable and family history unreliable or absent. Our previous screening of a selection of genes known to be involved in the pathogenesis of human cancers directly or indirectly according to the difference in the allelic burden in 690 MN patients (Li, S.T, Leukemia, 2020) we identified the myeloperoxidase (MPO) gene as having the highest burden of pathogenic alterations. MPO (17q22-23) encodes a heme-containing enzyme exclusively expressed in myeloid cells. MPO is known for its role in host defense against common microbial infections. In the presence of H2O2, MPO catalyzes the formation of hypohalous acids which are potent oxidants and strong bactericides. Many alterations in MPO have been found in hereditary MPO deficiency, an autosomal recessive syndrome with variable clinical penetrance. MPO deficiency syndrome is characterized by increased susceptibility to infections. To date, formal cancer association has not been established. In particular due to relatively mild phenotype, hereditary deficiency may remain unrecognized. Inspired by results of an initial screen we investigated MPO mutations in 3262 patients with various bone marrow failure syndromes (AA/PNH, 213) and MN (AML, 1655; MDS, 1133; MDS/MPN, 161; MPN, 100). In a subset of patients (20%), paired normal and tumor samples were sequenced in order to confirm germline origin. Germline MPO mutations were identified in 105 cases with 52% of them in AML. The most common recurrent mutation was c.2031-2A>C (42%; 44/105) (3' splice site of intron11) followed by I642L/F 15% (16/105), R569W 14 % (15/105), M519fs 13% (14/105), R460Q 7% (7/105), Y173C 5% (5/105), and R480H 2% (2/105). None of the variants were biallelic. The expected frequency of overall MPO variants in our cohort was higher than gnomAD (0.26% vs. 0.18%; P= .15×10-3) with odds ratio of 1.4. Notably, MPO mutations were significantly enriched in patients with age < 60 (2.2%; odds ratio =12.1, 95%CI=9.3-17.3; PC, R569W, M519fs, Y173C) contributed to this significant enrichment. Co-occurring lesions of germline MPO mutants (MPOMT) were then investigated: DNMT3A, SF3B1, TP53, NRAS were commonly observed in MPOMT compared to MPO WT-MN (20 vs. 14%, 17 vs. 13%, 16 vs. 8%, and 12 vs. 7%). Normal karyotype was found in 47% of carriers without enrichment for complex, -7/del(7q), del(5q), del(20q) or +8 karyotypes compared to WT. We compared RNA expression patterns between patients with pathogenic MPO mutations and those WT for MPO. Examination of genes significantly overexpressed in MPOMT showed strong enrichment in pseudoperoxidases hemoproteins involved in oxygen transport-globin (P=10-3) and heme biosynthesis (P=.012) (Fig.1C). The overexpression of hemoglobin/myoglobin genes in MPOMT suggests an excessive compensatory process due to the lack of true peroxidase activity. The fact that MPO has microbicidal activity led us to investigate the consequences of MPO mutations. Using shotgun whole-microbiome sequencing of circulating DNA, we examined the overall bacterial load in each patient. We found that MPO pathogenic mutation carriers had significantly higher bacterial burden than non-carriers (Fig.1D). The high bacterial burden of MPO mutants possibly promoted chronic inflammation, known to correlate with cancer pathogenesis. Indeed, we found that the presence of pathogenic MPO mutations was correlated with decreased survival in AML (P = 1.5 x 10-5). In sum, this is the first cohort showing germline MPO variants as predisposing alterations in MN. MPO variants might cause partial or total MPO deficiency resulting in chronic inflammatory responses by excessive compensation of immunity against invading microbes. This chronic inflammation stages can increase proliferation of HSC or myeloid progenitors and give rise to DNA replication associated mutations. Disclosures Maciejewski: Alexion, BMS: Speakers Bureau; Novartis, Roche: Consultancy, Honoraria.
- Published
- 2020
50. Somatic mosaicism in chronic myeloid leukemia in remission
- Author
-
Kenichi Chiba, Seishi Ogawa, Yuka Nakamura, Yukitsugu Nakamura, Motoshi Ichikawa, Yasunobu Nagata, Kenichi Yoshida, Ko Sasaki, Hiroko Tanaka, Yuichi Shiraishi, Kinuko Mitani, Satoru Miyano, and Hideki Makishima
- Subjects
Adult ,Male ,0301 basic medicine ,Immunology ,Fusion Proteins, bcr-abl ,Biology ,Biochemistry ,03 medical and health sciences ,Myelogenous ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Humans ,Protein Kinase Inhibitors ,neoplasms ,Myeloproliferative neoplasm ,Aged ,Aged, 80 and over ,Mosaicism ,Breakpoint ,Myeloid leukemia ,Hematopoietic stem cell ,Cell Biology ,Hematology ,Middle Aged ,medicine.disease ,Fusion protein ,Hematopoiesis ,Haematopoiesis ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Female - Abstract
To the editor: Chronic myeloid leukemia (CML) represents a distinct subtype of myeloproliferative neoplasm originated from a deregulated hematopoietic stem cell, in which a constitutively activated breakpoint cluster region-Abelson (BCR-ABL) fusion kinase plays a crucial role.[1][1] Although its
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.