Significance of Gene Diagnosis in Acute Myeloid Leukemia with the Emergence of New Molecular Target Drug Treatment

Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy accompanied by impaired differentiation and autonomous proliferation of hematopoietic stem cells. Standard induction therapy results in first complete remission among 70% of patients with AML; however, approximately half of the...

Full description

Saved in:
Bibliographic Details
Published inJournal of Nippon Medical School Vol. 89; no. 5; pp. 470 - 478
Main Author Yamaguchi, Hiroki
Format Journal Article
LanguageEnglish
Published The Medical Association of Nippon Medical School 01.01.2022
Subjects
Online AccessGet full text

Cover

Loading…
Abstract Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy accompanied by impaired differentiation and autonomous proliferation of hematopoietic stem cells. Standard induction therapy results in first complete remission among 70% of patients with AML; however, approximately half of these patients relapse and become refractory. Allogeneic hematopoietic cell transplantation is a useful treatment for relapsed and refractory cases. However, transplantation-related mortality is approximately 20%, which is not a low value, and quality of life after transplantation decreases. Therefore, there is a need to stratify the prognosis of each patient and implement this treatment appropriately. Owing to recent advances in genome analysis technology, many gene mutations involved in onset and recurrence of AML have been discovered. These abnormalities and mutations not only have clinical application as prognostic factors and minimal residual disease markers, but they may also contribute to novel molecular targeted drug development. Many new drugs such as first-generation FMS-like tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 and 2 (IDH1/2), and B cell lymphoma 2 (BCL2) inhibitors have been developed in the West. In addition, the second-generation FLT3 inhibitors gilteritinib and quizartinib were developed in Japan, and treatment outcomes for patients with AML have improved. However, there is still a large disparity in drug availability between the West, and Japan. As a result, treatment guidelines in the West cannot be applied in the clinical setting in Japan. In this study, we assessed the molecular target drug treatment by gene diagnosis for treatment of AML patients.
AbstractList Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy accompanied by impaired differentiation and autonomous proliferation of hematopoietic stem cells. Standard induction therapy results in first complete remission among 70% of patients with AML; however, approximately half of these patients relapse and become refractory. Allogeneic hematopoietic cell transplantation is a useful treatment for relapsed and refractory cases. However, transplantation-related mortality is approximately 20%, which is not a low value, and quality of life after transplantation decreases. Therefore, there is a need to stratify the prognosis of each patient and implement this treatment appropriately. Owing to recent advances in genome analysis technology, many gene mutations involved in onset and recurrence of AML have been discovered. These abnormalities and mutations not only have clinical application as prognostic factors and minimal residual disease markers, but they may also contribute to novel molecular targeted drug development. Many new drugs such as first-generation FMS-like tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 and 2 (IDH1/2), and B cell lymphoma 2 (BCL2) inhibitors have been developed in the West. In addition, the second-generation FLT3 inhibitors gilteritinib and quizartinib were developed in Japan, and treatment outcomes for patients with AML have improved. However, there is still a large disparity in drug availability between the West, and Japan. As a result, treatment guidelines in the West cannot be applied in the clinical setting in Japan. In this study, we assessed the molecular target drug treatment by gene diagnosis for treatment of AML patients.Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy accompanied by impaired differentiation and autonomous proliferation of hematopoietic stem cells. Standard induction therapy results in first complete remission among 70% of patients with AML; however, approximately half of these patients relapse and become refractory. Allogeneic hematopoietic cell transplantation is a useful treatment for relapsed and refractory cases. However, transplantation-related mortality is approximately 20%, which is not a low value, and quality of life after transplantation decreases. Therefore, there is a need to stratify the prognosis of each patient and implement this treatment appropriately. Owing to recent advances in genome analysis technology, many gene mutations involved in onset and recurrence of AML have been discovered. These abnormalities and mutations not only have clinical application as prognostic factors and minimal residual disease markers, but they may also contribute to novel molecular targeted drug development. Many new drugs such as first-generation FMS-like tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 and 2 (IDH1/2), and B cell lymphoma 2 (BCL2) inhibitors have been developed in the West. In addition, the second-generation FLT3 inhibitors gilteritinib and quizartinib were developed in Japan, and treatment outcomes for patients with AML have improved. However, there is still a large disparity in drug availability between the West, and Japan. As a result, treatment guidelines in the West cannot be applied in the clinical setting in Japan. In this study, we assessed the molecular target drug treatment by gene diagnosis for treatment of AML patients.
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy accompanied by impaired differentiation and autonomous proliferation of hematopoietic stem cells. Standard induction therapy results in first complete remission among 70% of patients with AML; however, approximately half of these patients relapse and become refractory. Allogeneic hematopoietic cell transplantation is a useful treatment for relapsed and refractory cases. However, transplantation-related mortality is approximately 20%, which is not a low value, and quality of life after transplantation decreases. Therefore, there is a need to stratify the prognosis of each patient and implement this treatment appropriately. Owing to recent advances in genome analysis technology, many gene mutations involved in onset and recurrence of AML have been discovered. These abnormalities and mutations not only have clinical application as prognostic factors and minimal residual disease markers, but they may also contribute to novel molecular targeted drug development. Many new drugs such as first-generation FMS-like tyrosine kinase 3 (FLT3), isocitrate dehydrogenase 1 and 2 (IDH1/2), and B cell lymphoma 2 (BCL2) inhibitors have been developed in the West. In addition, the second-generation FLT3 inhibitors gilteritinib and quizartinib were developed in Japan, and treatment outcomes for patients with AML have improved. However, there is still a large disparity in drug availability between the West, and Japan. As a result, treatment guidelines in the West cannot be applied in the clinical setting in Japan. In this study, we assessed the molecular target drug treatment by gene diagnosis for treatment of AML patients.
ArticleNumber JNMS.2022_89-313
Author Yamaguchi, Hiroki
Author_xml – sequence: 1
  fullname: Yamaguchi, Hiroki
  organization: Department of Hematology, Nippon Medical School
BookMark eNqFkE1v2zAMhoWhA9Z2-wkDdNzFmSxZUoydin5tbZIdmp0FRqEdZbbUSTKC_vs5TdBDLr2QBPg-JPBckDMfPBLytWSTkmv-fev7NHlYzJ8mnHFupnUhSvGBnJei0oWoWH32OsuiUlp9IhcpbRkTQkp1TuKTa71rnAVvkYaG3qNHeuOg9SG5RJ2nV3bISOcv2AW3pjMc_mLvgO5c3tC8QXrbY2zxiC9wR-ehQzt0EOkSxk2mN3Fo6TIi5B59_kw-NtAl_HLsl-TP3e3y-mcx-33_6_pqVlgpZC7WAoRtlGbA2UquLTClywq5rKSersRUy7pWFYxNr3SjGsa15RXK9aoua1txcUm-He4-x_BvwJRN75LFrgOPYUiGayFLpWopxqg8RG0MKUVszHN0PcQXUzKzd2z2js3esTk6NqPjkftxwlmXIbvgcwTXvUs_HuhtytDi20-I2dkOD9SYlftySr-l7AaiQS_-A2BEoco
CitedBy_id crossref_primary_10_1111_cas_15720
crossref_primary_10_1007_s12185_024_03824_x
crossref_primary_10_1007_s12185_024_03837_6
Cites_doi 10.1182/blood-2014-02-554667
10.1038/leu.2012.317
10.1182/blood.2019002697
10.1016/S1470-2045(14)70281-5
10.1200/JCO.2014.60.0890
10.1182/blood-2016-08-733196
10.1200/JCO.2006.06.9500
10.1038/leu.2011.104
10.1182/bloodadvances.2021005538
10.3390/ijms19113492
10.1046/j.1365-2141.2003.04362.x
10.1056/NEJMoa2012971
10.1038/s41375-020-0806-0
10.1016/S0140-6736(12)60485-1
10.1016/S1470-2045(19)30150-0
10.1056/NEJMoa1507471
10.1182/blood-2005-04-1466
10.1158/1078-0432.CCR-20-0834
10.1182/blood-2013-12-540971
10.1158/1078-0432.CCR-18-1897
10.1200/JCO.2017.77.6112
10.1016/j.bbmt.2016.03.015
10.1182/bloodadvances.2018020305
10.1007/s00277-017-3074-y
10.1056/NEJMoa2117344
10.1182/blood-2013-01-466706
10.1038/s41375-019-0472-2
10.1002/cncr.28974
10.1007/s12185-012-1150-6
10.1182/blood-2014-05-578070
10.1016/j.exphem.2017.05.003
10.1007/s12185-020-02894-x
10.1182/blood.2020004856
10.1182/blood-2012-06-431122
10.1056/NEJMoa1614359
10.3324/haematol.2016.143073
10.1056/NEJMoa1902688
10.1056/NEJMoa1005143
10.1182/bloodadvances.2021004292
10.1056/NEJMoa1716984
10.1007/s00277-020-04051-0
ContentType Journal Article
Copyright 2022 by the Medical Association of Nippon Medical School
Copyright_xml – notice: 2022 by the Medical Association of Nippon Medical School
DBID AAYXX
CITATION
7X8
DOI 10.1272/jnms.JNMS.2022_89-313
DatabaseName CrossRef
MEDLINE - Academic
DatabaseTitle CrossRef
MEDLINE - Academic
DatabaseTitleList MEDLINE - Academic

DeliveryMethod fulltext_linktorsrc
Discipline Medicine
EISSN 1347-3409
EndPage 478
ExternalDocumentID 10_1272_jnms_JNMS_2022_89_313
article_jnms_89_5_89_JNMS_2022_89_313_article_char_en
GroupedDBID .55
29L
2WC
53G
5GY
7.U
ABDBF
AEGXH
AENEX
ALMA_UNASSIGNED_HOLDINGS
CS3
DIK
DU5
E3Z
EBD
EBS
EJD
EMOBN
ESX
F5P
FRP
GX1
JMI
JSF
JSH
KQ8
MK0
MOJWN
M~E
RJT
RNS
RZJ
SV3
TR2
TUS
W2D
X7M
XSB
~8M
AAYXX
ACUHS
CITATION
OVT
7X8
ID FETCH-LOGICAL-c535t-d3a3cf670a20b5dca06714e254578b38759964a7597b7f6f027c24e5db919c423
ISSN 1345-4676
1347-3409
IngestDate Fri Jul 11 08:43:28 EDT 2025
Tue Jul 01 01:18:03 EDT 2025
Thu Apr 24 23:06:56 EDT 2025
Sun Jul 28 05:37:13 EDT 2024
IsDoiOpenAccess true
IsOpenAccess true
IsPeerReviewed true
IsScholarly true
Issue 5
Language English
LinkModel OpenURL
MergedId FETCHMERGED-LOGICAL-c535t-d3a3cf670a20b5dca06714e254578b38759964a7597b7f6f027c24e5db919c423
Notes ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
OpenAccessLink https://www.jstage.jst.go.jp/article/jnms/89/5/89_JNMS.2022_89-313/_article/-char/en
PQID 2735166953
PQPubID 23479
PageCount 9
ParticipantIDs proquest_miscellaneous_2735166953
crossref_primary_10_1272_jnms_JNMS_2022_89_313
crossref_citationtrail_10_1272_jnms_JNMS_2022_89_313
jstage_primary_article_jnms_89_5_89_JNMS_2022_89_313_article_char_en
ProviderPackageCode CITATION
AAYXX
PublicationCentury 2000
PublicationDate 2022-01-01
PublicationDateYYYYMMDD 2022-01-01
PublicationDate_xml – month: 01
  year: 2022
  text: 2022-01-01
  day: 01
PublicationDecade 2020
PublicationTitle Journal of Nippon Medical School
PublicationTitleAlternate J Nippon Med Sch
PublicationYear 2022
Publisher The Medical Association of Nippon Medical School
Publisher_xml – name: The Medical Association of Nippon Medical School
References 5. Kurosawa S, Yamaguchi H, Yamaguchi T, et al. The prognostic impact of FLT3-ITD, NPM1 and CEBPa in patients with intermediate-risk acute myeloid leukemia after first relapse. Int J Hematol. 2020 Aug;112 (2):200-9.
18. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363 (25):2424-33.
24. Wakita S, Yamaguchi H, Miyake K, et al. Importance of c-kit mutation detection method sensitivity in prognostic analyses of t (8;21) (q22;q22) acute myeloid leukemia. Leukemia. 2011;25 (9):1423-32.
27. Petersdorf SH, Kopecky KJ, Slovak M, et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood. 2013;121 (24):4854-60.
4. Kurosawa S, Yamaguchi H, Yamaguchi T, et al. Decision analysis of post-remission therapy in cytogenetically intermediate-risk AML: The impact of FLT3-ITD, NPM1, and CEBPA. Biol Blood Marrow Transplant. 2016;22 (6):1125-32.
9. Pratcorona M, Younis J, Moustafa H, et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121 (14):2734-8.
23. Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107 (5):1791-9.
33. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123 (21):3239-46.
13. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381 (18):1728-40.
1. Miyawaki S. Clinical studies of acute myeloid leukemia in the Japan Adult Leukemia Study Group. Int J Hematol. 2012;96 (2):171-7.
17. Wakita S, Yamaguchi H, Omori I, et al. Mutations of the epigenetics modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia. 2013;27 (5):1044-52.
29. Hills RK, Castaigne S, Appelbaum FR, et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15 (9):986-96.
39. Pollyea DA, Tallman MS, de Botton S, et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia. 2019;33 (11):2575-84.
3. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129 (4):424-47.
20. Herold T, Rothenberg-Thurley M, Grunwald VV, et al. Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia. Leukemia. 2020;34 (12):3161-72.
8. Linch DC, Hills RK, Burnett AK, et al. Impact of FLT3 (ITD) mutant allele level on relapse risk in intermediate-risk acute myeloid leukemia. Blood. 2014;124 (2):273-6.
16. Forghieri F, Comoli P, Marasca R, et al. Minimal/Measurable residual disease monitoring in NPM1-mutated acute myeloid leukemia: A clinical viewpoint and perspectives. Int J Mol Sci. 2018;19 (11):3492.
28. Castaigne S, Pautas C, Terré C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379 (9825):1508-16.
19. Ryotokuji T, Yamaguchi H, Ueki T, et al. Clinical characteristics and prognosis of acute myeloid leukemia associated with DNA-methylation regulatory gene mutations. Haemtologica. 2016;101 (9):1074-81.
14. Cortes JE, Khaled S, Martinelli G, et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019;20 (7):984-97.
37. Cherry EM, Abbott D, Amaya M, et al. Venetoclax and azacitidine compared with induction chemotherapy for newly diagnosed patients with acute myeloid leukemia. Blood Adv. 2021;5 (24):5565-73.
10. Sakaguchi M, Yamaguchi H, Najima Y, et al. Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv. 2018;2 (20):2744-54.
41. Norsworthy KJ, Mulkey F, Scott EC, et al. Differentiation syndrome with ivosidenib and enasidenib treatment in patients with relapsed or refractory IDH-mutated AML: A U.S. food and drug administration systematic analysis. Clin Cancer Res. 2020;26 (16):4280-8.
7. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124 (23):3441-9.
31. Granfeldt Østgård LS, Medeiros BC, Sengeløv H, et al. Epidemiology and clinical significance of secondary and therapy-related acute myeloid leukemia: A national population-based cohort study. J Clin Oncol. 2015;33 (31):3641-9.
32. Cortes JE, Goldberg SL, Feldman EJ, et al. Phase II, multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121 (2):234-42.
11. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377 (5):454-64.
21. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv (16) and t (8;21): a Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24 (24):3904-11.
30. Tarlock K, Alonzo TA, Wang YC, et al. Functional properties of KIT mutations are associated with differential clinical outcomes and response to targeted therapeutics in CBF acute myeloid leukemia. Clin Cancer Res. 2019;25 (16):5038-48.
34. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36 (26):2684-92.
35. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383 (7):617-29.
40. Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2022;386 (16):1519-31.
22. Care RS, Valk PJ, Goodeve AC, et al. Incidence and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias. Br J Haematol. 2003;121 (5):775-7.
36. Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020 Jun 11;135 (24):2137-45.
25. Yui S, Kurosawa S, Yamaguchi H, et al. D816 mutation of the KIT gene in core binding factor acute myeloid leukemia is associated with poorer prognosis than other KIT gene mutations. Ann Hematol. 2017;96:1641-52.
26. Omori I, Yamaguchi H, Miyake K, et al. D816V mutation in the KIT gene activation loop has a greater cell proliferative and anti-apoptotic ability than N822K mutation in core binding factor-acute myeloid leukemia. Exp Hematol. 2017;52:56-64.
2. Yanada M, Takami A, Yamasaki S, et al. Allogeneic hematopoietic cell transplantation for adults with acute myeloid leukemia conducted in Japan during the past quarter century. Ann Hematol. 2020;99 (6):1351-60.
6. Wakita S, Sakaguchi M, Oh I, et al. Prognostic impact of CEBPA bZIP domain mutation in acute myeloid leukemia. Blood Adv. 2022;6 (1):238-47.
38. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378 (25):2386-98.
12. Döhner K, Thiede C, Jahn N, et al. Impact of NPM1/FLT3-ITD genotypes defined by the2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood. 2019;135 (5):371-80.
15. Ivey A, Hills RK, Simpson MA, et al. Assessment of minimal residual disease in standard-risk AML. N Engl J Med. 2016 Feb 4;374 (5):422-33.
22
23
24
25
26
27
28
29
JE Cortes (32) 2015; 121
30
31
10
11
33
12
34
13
35
14
36
15
37
16
38
17
39
18
19
1
2
3
4
5
6
7
8
9
40
41
20
21
References_xml – reference: 22. Care RS, Valk PJ, Goodeve AC, et al. Incidence and prognosis of c-KIT and FLT3 mutations in core binding factor (CBF) acute myeloid leukaemias. Br J Haematol. 2003;121 (5):775-7.
– reference: 11. Stone RM, Mandrekar SJ, Sanford BL, et al. Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation. N Engl J Med. 2017;377 (5):454-64.
– reference: 24. Wakita S, Yamaguchi H, Miyake K, et al. Importance of c-kit mutation detection method sensitivity in prognostic analyses of t (8;21) (q22;q22) acute myeloid leukemia. Leukemia. 2011;25 (9):1423-32.
– reference: 21. Paschka P, Marcucci G, Ruppert AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv (16) and t (8;21): a Cancer and Leukemia Group B Study. J Clin Oncol. 2006;24 (24):3904-11.
– reference: 20. Herold T, Rothenberg-Thurley M, Grunwald VV, et al. Validation and refinement of the revised 2017 European LeukemiaNet genetic risk stratification of acute myeloid leukemia. Leukemia. 2020;34 (12):3161-72.
– reference: 38. DiNardo CD, Stein EM, de Botton S, et al. Durable remissions with ivosidenib in IDH1-mutated relapsed or refractory AML. N Engl J Med. 2018;378 (25):2386-98.
– reference: 25. Yui S, Kurosawa S, Yamaguchi H, et al. D816 mutation of the KIT gene in core binding factor acute myeloid leukemia is associated with poorer prognosis than other KIT gene mutations. Ann Hematol. 2017;96:1641-52.
– reference: 8. Linch DC, Hills RK, Burnett AK, et al. Impact of FLT3 (ITD) mutant allele level on relapse risk in intermediate-risk acute myeloid leukemia. Blood. 2014;124 (2):273-6.
– reference: 23. Schnittger S, Kohl TM, Haferlach T, et al. KIT-D816 mutations in AML1-ETO-positive AML are associated with impaired event-free and overall survival. Blood. 2006;107 (5):1791-9.
– reference: 2. Yanada M, Takami A, Yamasaki S, et al. Allogeneic hematopoietic cell transplantation for adults with acute myeloid leukemia conducted in Japan during the past quarter century. Ann Hematol. 2020;99 (6):1351-60.
– reference: 40. Montesinos P, Recher C, Vives S, et al. Ivosidenib and azacitidine in IDH1-mutated acute myeloid leukemia. N Engl J Med. 2022;386 (16):1519-31.
– reference: 3. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017;129 (4):424-47.
– reference: 39. Pollyea DA, Tallman MS, de Botton S, et al. Enasidenib, an inhibitor of mutant IDH2 proteins, induces durable remissions in older patients with newly diagnosed acute myeloid leukemia. Leukemia. 2019;33 (11):2575-84.
– reference: 16. Forghieri F, Comoli P, Marasca R, et al. Minimal/Measurable residual disease monitoring in NPM1-mutated acute myeloid leukemia: A clinical viewpoint and perspectives. Int J Mol Sci. 2018;19 (11):3492.
– reference: 27. Petersdorf SH, Kopecky KJ, Slovak M, et al. A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia. Blood. 2013;121 (24):4854-60.
– reference: 7. Schlenk RF, Kayser S, Bullinger L, et al. Differential impact of allelic ratio and insertion site in FLT3-ITD-positive AML with respect to allogeneic transplantation. Blood. 2014;124 (23):3441-9.
– reference: 9. Pratcorona M, Younis J, Moustafa H, et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121 (14):2734-8.
– reference: 18. Ley TJ, Ding L, Walter MJ, et al. DNMT3A mutations in acute myeloid leukemia. N Engl J Med. 2010;363 (25):2424-33.
– reference: 12. Döhner K, Thiede C, Jahn N, et al. Impact of NPM1/FLT3-ITD genotypes defined by the2017 European LeukemiaNet in patients with acute myeloid leukemia. Blood. 2019;135 (5):371-80.
– reference: 15. Ivey A, Hills RK, Simpson MA, et al. Assessment of minimal residual disease in standard-risk AML. N Engl J Med. 2016 Feb 4;374 (5):422-33.
– reference: 6. Wakita S, Sakaguchi M, Oh I, et al. Prognostic impact of CEBPA bZIP domain mutation in acute myeloid leukemia. Blood Adv. 2022;6 (1):238-47.
– reference: 31. Granfeldt Østgård LS, Medeiros BC, Sengeløv H, et al. Epidemiology and clinical significance of secondary and therapy-related acute myeloid leukemia: A national population-based cohort study. J Clin Oncol. 2015;33 (31):3641-9.
– reference: 29. Hills RK, Castaigne S, Appelbaum FR, et al. Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol. 2014;15 (9):986-96.
– reference: 34. Lancet JE, Uy GL, Cortes JE, et al. CPX-351 (cytarabine and daunorubicin) liposome for injection versus conventional cytarabine plus daunorubicin in older patients with newly diagnosed secondary acute myeloid leukemia. J Clin Oncol. 2018;36 (26):2684-92.
– reference: 30. Tarlock K, Alonzo TA, Wang YC, et al. Functional properties of KIT mutations are associated with differential clinical outcomes and response to targeted therapeutics in CBF acute myeloid leukemia. Clin Cancer Res. 2019;25 (16):5038-48.
– reference: 4. Kurosawa S, Yamaguchi H, Yamaguchi T, et al. Decision analysis of post-remission therapy in cytogenetically intermediate-risk AML: The impact of FLT3-ITD, NPM1, and CEBPA. Biol Blood Marrow Transplant. 2016;22 (6):1125-32.
– reference: 19. Ryotokuji T, Yamaguchi H, Ueki T, et al. Clinical characteristics and prognosis of acute myeloid leukemia associated with DNA-methylation regulatory gene mutations. Haemtologica. 2016;101 (9):1074-81.
– reference: 26. Omori I, Yamaguchi H, Miyake K, et al. D816V mutation in the KIT gene activation loop has a greater cell proliferative and anti-apoptotic ability than N822K mutation in core binding factor-acute myeloid leukemia. Exp Hematol. 2017;52:56-64.
– reference: 5. Kurosawa S, Yamaguchi H, Yamaguchi T, et al. The prognostic impact of FLT3-ITD, NPM1 and CEBPa in patients with intermediate-risk acute myeloid leukemia after first relapse. Int J Hematol. 2020 Aug;112 (2):200-9.
– reference: 13. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3-mutated AML. N Engl J Med. 2019;381 (18):1728-40.
– reference: 32. Cortes JE, Goldberg SL, Feldman EJ, et al. Phase II, multicenter, randomized trial of CPX-351 (cytarabine:daunorubicin) liposome injection versus intensive salvage therapy in adults with first relapse AML. Cancer. 2015;121 (2):234-42.
– reference: 14. Cortes JE, Khaled S, Martinelli G, et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): a multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019;20 (7):984-97.
– reference: 17. Wakita S, Yamaguchi H, Omori I, et al. Mutations of the epigenetics modifying gene (DNMT3a, TET2, IDH1/2) at diagnosis may induce FLT3-ITD at relapse in de novo acute myeloid leukemia. Leukemia. 2013;27 (5):1044-52.
– reference: 41. Norsworthy KJ, Mulkey F, Scott EC, et al. Differentiation syndrome with ivosidenib and enasidenib treatment in patients with relapsed or refractory IDH-mutated AML: A U.S. food and drug administration systematic analysis. Clin Cancer Res. 2020;26 (16):4280-8.
– reference: 28. Castaigne S, Pautas C, Terré C, et al. Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study. Lancet. 2012;379 (9825):1508-16.
– reference: 35. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383 (7):617-29.
– reference: 37. Cherry EM, Abbott D, Amaya M, et al. Venetoclax and azacitidine compared with induction chemotherapy for newly diagnosed patients with acute myeloid leukemia. Blood Adv. 2021;5 (24):5565-73.
– reference: 1. Miyawaki S. Clinical studies of acute myeloid leukemia in the Japan Adult Leukemia Study Group. Int J Hematol. 2012;96 (2):171-7.
– reference: 33. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a fixed 5:1 molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in older adults with untreated AML. Blood. 2014;123 (21):3239-46.
– reference: 36. Wei AH, Montesinos P, Ivanov V, et al. Venetoclax plus LDAC for newly diagnosed AML ineligible for intensive chemotherapy: a phase 3 randomized placebo-controlled trial. Blood. 2020 Jun 11;135 (24):2137-45.
– reference: 10. Sakaguchi M, Yamaguchi H, Najima Y, et al. Prognostic impact of low allelic ratio FLT3-ITD and NPM1 mutation in acute myeloid leukemia. Blood Adv. 2018;2 (20):2744-54.
– ident: 8
  doi: 10.1182/blood-2014-02-554667
– ident: 17
  doi: 10.1038/leu.2012.317
– ident: 12
  doi: 10.1182/blood.2019002697
– ident: 29
  doi: 10.1016/S1470-2045(14)70281-5
– ident: 31
  doi: 10.1200/JCO.2014.60.0890
– ident: 3
  doi: 10.1182/blood-2016-08-733196
– ident: 21
  doi: 10.1200/JCO.2006.06.9500
– ident: 24
  doi: 10.1038/leu.2011.104
– ident: 37
  doi: 10.1182/bloodadvances.2021005538
– ident: 16
  doi: 10.3390/ijms19113492
– ident: 22
  doi: 10.1046/j.1365-2141.2003.04362.x
– ident: 35
  doi: 10.1056/NEJMoa2012971
– ident: 20
  doi: 10.1038/s41375-020-0806-0
– ident: 28
  doi: 10.1016/S0140-6736(12)60485-1
– ident: 14
  doi: 10.1016/S1470-2045(19)30150-0
– ident: 15
  doi: 10.1056/NEJMoa1507471
– ident: 23
  doi: 10.1182/blood-2005-04-1466
– ident: 41
  doi: 10.1158/1078-0432.CCR-20-0834
– ident: 33
  doi: 10.1182/blood-2013-12-540971
– ident: 30
  doi: 10.1158/1078-0432.CCR-18-1897
– ident: 34
  doi: 10.1200/JCO.2017.77.6112
– ident: 4
  doi: 10.1016/j.bbmt.2016.03.015
– ident: 10
  doi: 10.1182/bloodadvances.2018020305
– ident: 25
  doi: 10.1007/s00277-017-3074-y
– ident: 40
  doi: 10.1056/NEJMoa2117344
– ident: 27
  doi: 10.1182/blood-2013-01-466706
– ident: 39
  doi: 10.1038/s41375-019-0472-2
– volume: 121
  start-page: 234
  issn: 0008-543X
  issue: 2
  year: 2015
  ident: 32
  publication-title: Cancer
  doi: 10.1002/cncr.28974
– ident: 1
  doi: 10.1007/s12185-012-1150-6
– ident: 7
  doi: 10.1182/blood-2014-05-578070
– ident: 26
  doi: 10.1016/j.exphem.2017.05.003
– ident: 5
  doi: 10.1007/s12185-020-02894-x
– ident: 36
  doi: 10.1182/blood.2020004856
– ident: 9
  doi: 10.1182/blood-2012-06-431122
– ident: 11
  doi: 10.1056/NEJMoa1614359
– ident: 19
  doi: 10.3324/haematol.2016.143073
– ident: 13
  doi: 10.1056/NEJMoa1902688
– ident: 18
  doi: 10.1056/NEJMoa1005143
– ident: 6
  doi: 10.1182/bloodadvances.2021004292
– ident: 38
  doi: 10.1056/NEJMoa1716984
– ident: 2
  doi: 10.1007/s00277-020-04051-0
SSID ssj0033556
Score 2.2654011
SecondaryResourceType review_article
Snippet Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy accompanied by impaired differentiation and autonomous proliferation of hematopoietic...
SourceID proquest
crossref
jstage
SourceType Aggregation Database
Enrichment Source
Index Database
Publisher
StartPage 470
SubjectTerms acute myeloid leukemia
allogeneic hematopoietic cell transplantation
FLT3 inhibitor
gene diagnosis
molecular target drug treatment
Title Significance of Gene Diagnosis in Acute Myeloid Leukemia with the Emergence of New Molecular Target Drug Treatment
URI https://www.jstage.jst.go.jp/article/jnms/89/5/89_JNMS.2022_89-313/_article/-char/en
https://www.proquest.com/docview/2735166953
Volume 89
hasFullText 1
inHoldings 1
isFullTextHit
isPrint
ispartofPNX Journal of Nippon Medical School, 2022/10/25, Vol.89(5), pp.470-478
link http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1Lj9MwELZgQYgL4inKS0aCU5XSOk7SHFe0q2XVlsOmUm-W4zgl221SdVsh9tfzOXHSwhaxcEkr146rmc_jGXsehHxQSRyonq-dlEvf4UEcOGHCuaO7EpsJVnqsTHDyeOKfTvnZzJvt0hOU0SWbuKOuD8aV_A9X0Qa-mijZf-Bs81I04Dv4iyc4jOeteHyezXPj6lO6_UPrMzmkIcRK57msdHQ9VsYPYPxDXxZZ0h7p7UIvM2lPX4GQoY2-rE4O9HescVsutx2VPuLtwXo7b0e1O_ofdNlJtloVu1sfm9rzozuof_mykABYNpeLbfsa-vq3rBE3cinnpiJLuQlm62KR7Z9EMLZ3ElEJT5dDYPFuJQL1gTYrcauiQRZZ3p745FURkRtinQUmTexFvrzqnE3G5x0zu-iH2EDc3T5W391PvoqT6WgkouEsukvuMdgPZRT4rPH9caFklWFn9Z-zoV2Y5tPBSX5RWu5fQG-f39y8S40kekweWfLT4woXT8gdnT8lD8bWWeIZWe_DgxYpNfCgDTxoltMSHtTCg9bwoAYeFPCgDTzMcMCDNvCgFTyogQdt4PGcTE-G0edTx5bYcJTnehsncaWrUj_AyuzGXqIklJce1wx6ddCPXRizsIe5xAdWceqnXRYoxrWXxGEvVFDFX5CjvMj1S0JhGYQhZ0ymrsdhVYQhS1lfxSGYyvtMtgivaSiUzT9vyqBcCmOHgvTCkF4Y0gtLegHSt0inGbaqErD8bcCgYlDT3a7Pqjs6eebx-7Cml4l2hHBpkfc1ewUkrrlGk7kutlcCCr_X8_3Qc1_dos9r8nC3Vt6Qo816q99Cj93E70pM_gQx95_w
linkProvider Geneva Foundation for Medical Education and Research
openUrl ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Significance+of+Gene+Diagnosis+in+Acute+Myeloid+Leukemia+with+the+Emergence+of+New+Molecular+Target+Drug+Treatment&rft.jtitle=Journal+of+Nippon+Medical+School+%3D+Nippon+Ika+Daigaku+zasshi&rft.au=Yamaguchi%2C+Hiroki&rft.date=2022-01-01&rft.issn=1347-3409&rft.eissn=1347-3409&rft.volume=89&rft.issue=5&rft.spage=470&rft_id=info:doi/10.1272%2Fjnms.JNMS.2022_89-313&rft.externalDBID=NO_FULL_TEXT
thumbnail_l http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=1345-4676&client=summon
thumbnail_m http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=1345-4676&client=summon
thumbnail_s http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=1345-4676&client=summon