Next Article in Journal
The Impact of Core Tissues on Successful Next-Generation Sequencing Analysis of Specimens Obtained through Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration
Previous Article in Journal
The HROC-Xenobank—A High Quality Assured PDX Biobank of >100 Individual Colorectal Cancer Models
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Comment

Comment on Geoffroy, M.-C.; de Thé, H. Classic and Variants APLs, as Viewed from a Therapy Response. Cancers 2020, 12, 967

1
Department of Haematology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
2
Department of Stem Cell Transplantation, Blood Diseases Hospital & Institute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(23), 5883; https://doi.org/10.3390/cancers13235883
Submission received: 13 September 2021 / Revised: 31 October 2021 / Accepted: 5 November 2021 / Published: 23 November 2021
Acute promyelocytic leukemia (APL) is a unique and very deeply studied acute myeloid leukemia. The typical molecular genetic feature of APL is the rearrangement of RARA gene with PML and a few with other partner genes. All-trans retinoic acids and arsenic trioxides are effective agents against APL harboring PML-RARA. However, there are still some rare cases lacking RARA fusion although they share the same morphology and immunocytochemistry features with typical APL. The classification of such cases remains controversial. They are usually referred to as acute promyelocytic-like leukemias (APLL), and sometimes they were morphologically classified as M3 according to the FAB classification. They are not regarded as APL in the WHO’s classification of myeloid neoplasms and acute leukemia. For years, the underlying molecular pathology of APL-like leukemia has remained concealed. Some researchers supposed that APL-like leukemia might bear genetic mutation of other members of retinoic acid receptors (RARs), such as RARB or RARG [1].
Due to the limitations of technology, identification of unknown fusion genes has been a relatively difficult task. In recent years, the emerging high-throughput sequencing technologies have provided powerful research tools for the discovery of novel fusion genes. In 2011, a novel NUP98-RARG fusion gene was discovered by Such E et al. in a Spanish case of APL-like leukemia, and it was the first reported fusion gene of this kind of leukemia. Since 2017, several other novel fusion genes harboring RARG have been found. Geoffroy et al. made a relatively comprehensive review in this field. In this review, a total of four RARG-related fusion genes are summarized, namely CPSF6-RARG & RARG-CPSF6, NUP98-RARG, PML-RARG, and NPM1-RARG-NPM1. NUP98-RARG accounts for a total of three cases and CPSF6-RARG & RARG-CPSF6 for five cases. CPSF6-RARG & RARG-CPSF6 own the most variants, consisting of four types of CPSF6-RARG and one type of RARG-CPSF6 [2]. In fact, another patient with CPSF6-RARG has been reported in 2019 [3]. Since then, two more cases of CPSF6-RARG and two cases of NUP98-RARG have been reported [4,5,6,7]. Therefore, the number of reported patients has reached eight cases for CPSF6-RARG and six for NUP98-RARG. Their biological and clinical information are summarized in Table 1 and Table 2, respectively.
Actually, RARG-related fusion genes that have been identified were not merely the four types mentioned. Su et al., reported a 43-year-old man with leukocytosis, anemia, and thrombocytopenia. Similarly, the morphology and immunological characteristics of the blasts were the same as APL. Transcriptome sequencing identified a new HNRNPC-RARG fusion gene and its reciprocal. The patient received ATRA induction therapy for three weeks, but the bone marrow blasts were almost unchanged. Then, combined chemotherapy (homoharringtonine, daunorubicin and cytarabine, HAD) was applied and complete remission was achieved. After that, he was given several courses of consolidation and intensive chemotherapy consecutively, although he relapsed one year later. Re-induction therapy medications included ATRA, arsenic trioxide, and combination chemotherapy, which, as inu other such cases, have not worked [16].
The RARG gene is located at chromosome 12q13. Several more APL-like cases with chromosome translocation involving 12q13 have been reported in previous pieces of literature, but further molecular detections were not performed (Table 3). Due to the fact that this kind of disease is relatively rare and due to the current limitations of research conditions, there may be more such cases unpublished. Actually, we also know that this situation does exist. As exhibited in Table 3, most cases are t(11;12) translocations, which are consistent with the location of the NUP98-RARG fusion gene. One of them is t(4;12) translocation, suggesting that there may be undiscovered new fusion genes. These cases also did not respond to the treatment of retinoic acid or arsenic trioxide.
Interestingly, there is another group of AML with t(11;12)(p15;q13), of which the cell morphology is not promyelocyte type, but classified into M1, M2, M4, and M5 according to FAB classification. In the fusion gene of such cases, one partner gene is also NUP98 located on chromosome 11p15, and the other is one of HOXC (HOXC11, HOXC13) gene clusters [18,21]. It should be noted that a series of genes are densely distributed in the region of human chromosome 12q13, including RARG and HOXC.
All reported cases of APLL with 12q13 rearrangement achieved no response to ATRA or arsenic trioxide treatment in vivo. In vitro culture of bone marrow blast cells from the first patient with NUP98-RARG also confirmed the failure, although hematopoietic stem cells transfected with NUP98-RARG were demonstrated to be sensitive to ATRA. Some researchers attributed this contradiction to the influence of the in vitro culture environment or additional mutations [22]. In fact, a series of RARs modulators have been synthesized, some of which are pan-antagonists/agonists and some of which are selective ones. ATRA is one of pan- agonists [23]. Researchers have observed that BMS961 (an RARG agonist) has not shown any effect on leukemia cells with RARG-CPSF6 cultured in vitro [10].
In most of the published reports and WHO classification, as mentioned earlier, APLL is regarded more as a subclass of AML than as an APL. However, the authors of this review pointed out in another article [24] that RARB and RARG are also retinoic acid receptors and play a key role in leukemogenesis through their rearrangement (similar to RARA). Moreover, APLL carrying RARG-CPSF6 shared the same gene expression profiles with classical APL harboring PML-RARA [10]. Although APLL is not responsive to ATRA, new target drugs (e.g., other RARs modulators) may be explored to improve the outcome of APLL patients. Thus, we think it seems to be reasonable to classify the APL and APLL into a same subclass and the APL with RARs rearrangements.
However, there have existed cases of APLL lacking RARs fusion genes. Researchers further explored other possible molecular abnormalities (e.g., dysregulation of RARA and RARG has been reported) [25,26]. Zhao et al., identified two new fusion genes NPM1-CCDC28A and TBC1D15-RAB21 in two cases of children without RARs rearrangement. The abnormal chromosome of the third patient was t(1;11)(q21;q23), and DNA sequencing identified two fusion genes, KMT2A-MLLT11 and RPRD2-KMT2A [3]. In addition, sporadic APLL literature could be retrieved which did not carry out in-depth study of molecular pathology, and some involved known genetic aberration such as JAK2 V617F mutation [27].
In conclusion, the identification of RARG/RARB related fusion genes over the last decade has revealed the mystery of APLL, and it can be predicted that more cases will be reported in the future. However, due to the insensitivity to ATRA and arsenite acid, there is still a lack of effective targeted drugs. In addition, there are some cases lacking RARs rearrangements, of which the molecular pathology remains to be clarified.

Author Contributions

Z.S. and X.L. contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marinelli, A.; Bossi, D.; Pelicci, P.G.; Minucci, S. Redundant function of retinoic acid receptor isoforms in leukemogenesis unravels a prominent function of genome topology and architecture in the selection of mutagenic events in cancer. Leukemia 2009, 23, 417–419. [Google Scholar] [CrossRef] [PubMed]
  2. Geoffroy, M.-C.; de Thé, H. Classic and Variants APLs, as Viewed from a Therapy Response. Cancers 2020, 12, 967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zhao, J.; Liang, J.-W.; Xue, H.-L.; Shen, S.-H.; Chen, J.; Tang, Y.-J.; Yu, L.-S.; Liang, H.-H.; Gu, L.-J.; Tang, J.-Y.; et al. The genetics and clinical characteristics of children morphologically diagnosed as acute promyelocytic leukemia. Leukemia 2019, 33, 1387–1399. [Google Scholar] [CrossRef] [PubMed]
  4. Tao, S.; Song, L.; Deng, Y.; Chen, Y.; Shi, Y.; Gan, Y.; Deng, Z.; Ding, B.; He, Z.; Wang, C.; et al. Acute Myeloid Leukemia with NUP98-RARG Gene Fusion Similar to Acute Promyelocytic Leukemia: Case Report and Literature Review. OncoTargets Ther. 2020, 13, 10559–10566. [Google Scholar] [CrossRef] [PubMed]
  5. Wei, W.; Liu, Q.; Song, F.; Cao, H.; Liu, M.; Jiang, Y.; Li, Y.; Gao, S. Alkaloid-based regimen is beneficial for acute myeloid leukemia resembling acute promyelocytic leukemia with NUP98/RARG fusion and RUNX1 mutation: A case report. Medicine 2020, 99, e22488. [Google Scholar] [CrossRef] [PubMed]
  6. Han, X.; Jin, C.; Zheng, G.; Li, Y.; Wang, Y.; Zhang, E.; Zhu, H.; Cai, Z. Acute myeloid leukemia with CPSF6-RARG fusion resembling acute promyelocytic leukemia with extramedullary infiltration. Ther. Adv. Hematol. 2021, 12, 2040620720976984. [Google Scholar] [CrossRef] [PubMed]
  7. Jiang, M.; Zhou, Y.R.; Zhan, Y.; Zhang, H.Q.; Zhang, Q.; Guo, Y.; Zhang, Z.L. Application of transcriptome sequencing and fusion genes analysis in the diagnosis of myeloid leukemia with normal karyotype. Zhonghua Yi Xue Za Zhi 2021, 101, 939–944. [Google Scholar]
  8. Liu, T.; Wen, L.; Yuan, H.; Wang, Y.; Yao, L.; Xu, Y.; Cen, J.; Ruan, C.; Wu, D.; Chen, S. Identification of novel recurrent CPSF6-RARG fusions in acute myeloid leukemia resembling acute promyelocytic leukemia. Blood 2018, 131, 1870–1873. [Google Scholar] [CrossRef]
  9. Qin, Y.-Z.; Huang, X.-J.; Zhu, H.-H. Identification of a novel CPSF6-RARG fusion transcript in acute myeloid leukemia resembling acute promyelocytic leukemia. Leukemia 2018, 32, 2285–2287. [Google Scholar] [CrossRef]
  10. Miller, C.A.; Tricarico, C.; Skidmore, Z.L.; Uy, G.L.; Lee, Y.-S.; Hassan, A.; O’Laughlin, M.D.; Schmidt, H.; Tian, L.; Duncavage, E.J.; et al. A case of acute myeloid leukemia with promyelocytic features characterized by expression of a novel RARG-CPSF6 fusion. Blood Adv. 2018, 2, 1295–1299. [Google Scholar] [CrossRef]
  11. Zhang, Z.; Jiang, M.; Borthakur, G.; Luan, S.; Huang, X.; Tang, G.; Xu, Q.; Ji, D.; Boyer, A.D.; Li, F.; et al. Acute myeloid leukemia with a novel CPSF6-RARG variant is sensitive to homoharringtonine and cytarabine chemotherapy. Am. J. Hematol. 2020, 95, E48–E51. [Google Scholar] [CrossRef]
  12. Such, E.; Cervera, J.; Valencia, A.; Barragán, E.; Ibañez, M.; Luna, I.; Fuster, O.; Perez-Sirvent, M.L.; Senent, L.; Sempere, A.; et al. A novel NUP98/RARG gene fusion in acute myeloid leukemia resembling acute promyelocytic leukemia. Blood 2011, 117, 242–245. [Google Scholar] [CrossRef]
  13. Such, E.; Cordón, L.; Sempere, A.; Villamón, E.; Ibañez, M.; Luna, I.; Gómez-Seguí, I.; López-Pavía, M.; Alonso, C.; Lo-Coco, F.; et al. In vitro all-trans retinoic acid sensitivity of acute myeloid leukemia blasts with NUP98/RARG fusion gene. Ann. Hematol. 2014, 93, 1931–1933. [Google Scholar] [CrossRef]
  14. Luo, H.; Zhang, S.; Li, K.; Chen, X.-H.; Li, Y.-C.; Sun, Y.; Liu, L.-F.; Yu, H.-Y.; Zhu, H.-H. A novel entity of acute myeloid leukaemia with recurrent RARG-rearrangement resembling acute promyelocytic leukaemia. Leuk. Res. 2019, 77, 14–16. [Google Scholar] [CrossRef]
  15. Zhang, X.; Li, F.; Wang, J.; Suo, S.; Ling, Q.; Yu, W.; Jin, J. RARγ-rearrangements resemble acute promyelocytic leukemia and benefit from 3 + 7 regimen. Leuk. Lymphoma 2019, 60, 1831–1834. [Google Scholar] [CrossRef]
  16. Su, Z.; Liu, X.; Xu, Y.; Hu, W.; Zhao, C.; Zhao, H.; Feng, X.; Zhang, S.; Yang, J.; Shi, X.; et al. Novel reciprocal fusion genes involving HNRNPC and RARG in acute promyelocytic leukemia lacking RARA rearrangement. Haematologica 2020, 105, e376–e378. [Google Scholar] [CrossRef]
  17. Qiu, H.; Xue, Y.; Pan, J.; Wu, Y.; Wang, Y.; Shen, J.; Zhang, J. A novel t(4; 12) (q11; q13) in a patient with acute myeloid leukemia. Cancer Genet. Cytogenet. 2007, 172, 176–177. [Google Scholar] [CrossRef]
  18. Gong, B.-F.; Li, Q.-H.; Li, W.; Wang, Y.; Wei, H.; Wang, J.-Y.; Zhao, X.-L.; Lin, D.; Li, C.-W.; Liu, X.-P.; et al. Acute myeloid leukemia with t(11; 12)(p15; q13) translocation: Two cases report and literature review. Zhonghua Xue Ye Xue Za Zhi 2013, 34, 830–833. [Google Scholar]
  19. Hua, J.; Bao, X.; Xie, Y. A Rare Morphology Resembling APL with t(11; 12) (p15; q13) in Acute Myeloid Leukemia: Case Report and Literature Review. Clin. Lab. 2019, 65. [Google Scholar] [CrossRef]
  20. Su, Y.; Liu, W.; Guan, Y.; Song, L.; Xie, X. Clinical observation of acute myeloid leukemia with morphology supporting the diagnosis of acute promyelocytic leukemia and PML/RARa gene negative. Mod. Med. J. China 2019, 21, 42–44. [Google Scholar]
  21. Hu, J.; Hong, X.; Li, Z.; Lu, Q. Acute monocytic leukaemia with t(11; 12) (p15; q13) chromosomal changes: A case report and literature review. Oncol. Lett. 2015, 10, 2307–2310. [Google Scholar] [CrossRef] [Green Version]
  22. Chlapek, P.; Slavikova, V.; Mazanek, P.; Sterba, J.; Veselska, R. Why Differentiation Therapy Sometimes Fails: Molecular Mechanisms of Resistance to Retinoids. Int. J. Mol. Sci. 2018, 19, 132. [Google Scholar] [CrossRef] [Green Version]
  23. Mendoza-Parra, M.A.; Bourguet, W.; de Lera, A.R.; Gronemeyer, H. Retinoid Receptor-Selective Modulators. In The Retinoids: Biology, Biochemistry, and Disease; Dollé, P., Niederreither, K., Eds.; John Wiley & Sons: Hoboken, NJ, USA, 2015; pp. 165–192. ISBN 9781118628003. [Google Scholar]
  24. Geoffroy, M.-C.; Esnault, C.; de Thé, H. Retinoids in hematology: A timely revival? Blood 2021, 137, 2429–2437. [Google Scholar] [CrossRef]
  25. Coccaro, N.; Zagaria, A.; Orsini, P.; Anelli, L.; Tota, G.; Casieri, P.; Impera, L.; Minervini, A.; Minervini, C.F.; Cumbo, C.; et al. RARA and RARG gene downregulation associated with EZH2 mutation in acute promyelocytic-like morphology leukemia. Hum. Pathol. 2018, 80, 82–86. [Google Scholar] [CrossRef]
  26. Zhang, X.; Huang, X.; Xu, H.; Li, J.; Yu, W. MLL-rearrangement can resemble acute promyelocytic leukemia. Leuk. Lymphoma 2019, 60, 2841–2843. [Google Scholar] [CrossRef]
  27. Forghieri, F.; Morselli, M.; Potenza, L.; Maccaferri, M.; Pedrazzi, L.; Coluccio, V.; Barozzi, P.; Vallerini, D.; Riva, G.; Zanetti, E.; et al. A case of JAK2 V617F-positive myelodysplastic/myeloproliferative neoplasm with unusual morphology, resembling acute promyelocytic leukemia-like disorder with a chronic course. Leuk. Lymphoma 2011, 52, 2012–2019. [Google Scholar] [CrossRef]
Table 1. Cases of APLL harboring CPSF6-RARG or RARG-CPSF6.
Table 1. Cases of APLL harboring CPSF6-RARG or RARG-CPSF6.
No.Age/
Gender
Promyelocytes PercentageCytogenetics and Molecular AberrationsImmunophenotypingATRA/ATO
Responsivity
Treatment and EfficacyReferences
148/F89%92, XXXX (2);
CPSF6-RARG;
DNMT3A-G587fs mutation
Positive: CD13, CD33, and MPO; partially positive: CD9, CD64; negative: HLA-DR, CD117, CD34, CD14 and CD11bATRA+ATO, noneATRA+ATO+idarubicin, ATRA+ATO+IA+G-CSF, then decitabine, NR. Abondoned treatment then died[8]
251/F87.5%del(12)(p12)(2)/46,XX (18);
CPSF6-RARG;
WT1 and K-RAS mutations
Positive: CD13, CD33, MPO, and CD9; partially positive: CD34; negative: HLA-DR, CD2, CD7, CD10, CD11c, CD14, and CD38ATRA, noneATRA+daunorubicin, NR; then DA, morphologic remission. followed by 2 courses of HD-AC and 2 courses of 7+3 chemotherapy, CR[8]
338/M65%46,XY (20);
CPSF6-RARG;
WT1-R462Q mutations
Positive: CD117, CD123, CD34, CD33 and CD13; partially positive: CD9, CD64; negative: CD11b, HLA-DR, CD38, CD56, and CD14ATRA, none. suspected differentiation syndromeATRA+RIF, then MA as induction therapy, died on the 37th day[9]
426/M60% blasts, 15% promyelocytes45,X,-Y (10)/45, idem, add(6)(q?13)(2)/46,XY (8);
RARG-CPSF6;
BMPR1A, NEAT1, WT1 mutations
Positive: CD33, CD13, CD64; partially positive: CD117; weak: HLA-DR; negative: CD34, CD56, CD19, CD2, CD5, CD123, CD14, CD11b, and TdTATRA, not exhibitedATRA+IA, lacking efficacy introduction[10]
55/MNot exhibited46,XY;
RARG-CPSF6/CPSF6-RARG
Not exhibitedNot exhibitedATRA+chemotherapy, relapse then death(11 months from diagnosis)[3]
655/M93%46,XY;
RARG-CPSF6
Positive: CD13, CD33, CD117, CD56; negative: HLA-DR, CD34, CD38, CD15, CD14, CD7, CD2, CD3, CD4, CD8, CD19, CD20, CD10ATRA+ATO, noneATRA+ATO, switched to IA, NR; then HA, CR. followed by 4 courses of HA and 1 course of EA as maintenance therapy[11]
767/F72%46,XX;
CPSF6-RARG;
WT1 mutation
Positive: MPO, CD13, CD33; partially positive: CD71, negative: CD14, CD19, CD34, CD38, CD64, CD117, CD11b, CD11c, HLA-DRATRA(10 days), NR1 week of ATRA then plus HA, died during the course[6]
855/M5.5% blasts, 88% promyelocytes46,XY;
CPSF6-RARG
Positive: CD33, CD13, CD117, CD56, negative: CD34, HLA-DRATRA, noneATRA plus 3 + 7 schedule, NR; then HA, sustained CR[7]
No., number; ATRA, all-trans retinoic acid; ATO, arsenic trioxide; M, male; F, female; RIF, oral arsenic realgar-indigo naturalis formula; IA, idarubicin and cytarabine; DA, Daunorubicin and cytarabine; HD-AC, high dose cytarabine; MA, mitoxantrone and cytarabine; HA, homoharringtonine and cytarabine; EA, etoposide and cytarabine; NR, no response; CR, complete response.
Table 2. Cases of APLL harboring NUP98-RARG.
Table 2. Cases of APLL harboring NUP98-RARG.
No.Age/
Gender
Promyelocytes PercentageCytogenetics and MutationImmunophenotypingATRA/ATO
Responsivity
Treatment and EfficacyReferences
135/M80%46,XY,t(11;12)(p15;q13) (16)/46,XY (4)Positive: CD13, CD33, CD45, CD117, cMPO; weakly positive: CD34; negative: HLA-DR, B or T-cell markersATRA, unknown in vivo, none in vitroIA, CR, followed by consolidation chemotherapy, then auto-PBSCT, CR for 2 years. Died when relapsed during ATRA+salvage treatment+UCBT[12,13]
245/F94.5%46,XX,t(11;12)(p15;q13) (16)/46 XX (4);
WT1-R445W mutation
Positive: MPO, CD117, CD33, CD13, CD38, CD64; negative: CD34, CD11b, HLA-DR, CD56, CD14, B or T-cell markersNoneATRA+ATO, switched to CA, NR and died[14]
322/M91%46,XY,t(11;12)(p15;q13);
WT1(c.1255_1256insGG, c643C>T) mutations
Positive: CD117, CD13, CD33, partially positive: HLA-DR; negative: CD34NoneATRA+ATO+ idarubicin, then HAA, NR; switched to DA, PR; followed by DA, CR[15]
447/F96.5%45,X,–X, del(9)(q13q22), t(11;12)(p15;q13) (20);
IDH2, TET2, ASXL1, TP53, WT1(exon7, exon9) mutations
Positive: MPO, CD13, CD33, HLA-DR, CD56ATRA+ATO (14 days), noneATRA+ATO, IA, HIAG, sequentially, CR; then HIAG, 2 cycles of half-CAG, 2 cycles of HA as consolidation chemotherapy; leukemia-free in 24-month follow-up[4]
518/M90.5%46,XY;
RUNX1:c.319C>A mutation(onset), 8 point mutations in WT1(relapse)
Positive: CD117, CD13, CD33, CD9, CD64, CD123, cMPO; negative: HLA-DR, CD34, CD38, CD11b, B or T-cell markersATRA (5 days)+ ATO (13 days), NRATRA+ATO, switched to DA, NR; then HAA, PR; HAA, HD-AC, CR; the patient abandoned further treatment and relapsed 3 months later and died[5]
633/M93%46,XYPositive: CD33, CD13, CD117; negative: CD34, HLA-DRNoneATRA plus 3 + 7 schedule, NR and died[7]
No., number; ATRA, all-trans retinoic acid; ATO, arsenic trioxide; M, male; F, female; auto-PBSCT, autologous peripheral blood stem-cell transplant; IA, idarubicin and cytarabine; UCBT, umbilical cord blood transplantation; CA, aclamycin and cytarabine; HAA, homoharringtonine, aclarubicin, and cytarabine; DA, daunorubicin and cytarabine; HIAG, homoharringtonine, idarubicin, cytarabine, and granulocyte colony-stimulating factor(GCSF); half-CAG, half-dose aclarubicin, cytarabine, and GCSF; HA, homoharringtonine and cytarabine; HD-AC, high dose cytarabine; NR, no response; CR, complete response; PR, partial response.
Table 3. Cases of APLL with 12q13 rearrangement and unidentified fusion gene.
Table 3. Cases of APLL with 12q13 rearrangement and unidentified fusion gene.
No.Age/
Gender
Promyelocytes PercentageCytogenetics and MutationImmunophenotypingATRA/ATO
Responsivity
Treatment and EfficacyReferences
137/M87%46,XY,t(4;12)(q11;q13) (29)/46,XY (6)Positive: CD13, CD33, negative: CD34, HLA-DRATO (15 days), NRATO+CAG as induction therapy, died during the course[17]
214/M90%46,XY,t(11;12)(p15;q13)Positive: CD13, CD33, CD 14, CD 64, CD34ATRA (2 weeks), NR; ATO (1 month), noneATRA followed by ATO, then DA, MA as induction therapy, NR and died[18]
351/M81.5%48,X Y,t(11;12) (p15;q13),+14,+21 (10)/46,XY (10);
FLT3-T K D mutation
Positive: CD13, CD33; partially positive: CD4, CD15, CD34, CD38, CD64, CD117; minority positive: CD11b; negative: HLA-DRATRA (8 days), NR, ATO (21 days), noneATRA+ATO, then HDA as induction therapy, CR; followed by D+ID-AC, M+ID-AC, HA, HA, MA, MA as consolidation therapy[18]
421/M89%46,XY,t(11;12)(p15;q13) (20)Positive: CD117, CD33, CD13; partially positive: CD64; negative: CD34, CD14, CD56, CD4, CD19, HLA-DRNoneATRA+ATO+idarubicin, HAA, NR; then IA, CR; died after consolidation therapy[19]
560/M85.6%46,XY,t(11;12)(p15;q13)Positive: cMPO, CD13, CD33, CD117, CD64, negative: CD34, HLA-DRNoneATRA+ATO, NR; then DA, CR[20]
649/M78%46,XY,t(11;12)(p15;q13)Positive: cMPO, CD13, CD33, CD117, CD64, negative: CD34, HLA-DRNoneATRA+ATO, NR; then HAA, CR[20]
No., number; ATRA, all-trans retinoic acid; ATO, arsenic trioxide; M, male; F, female; CAG, aclamycin, cytosine arabinoside, and granulocyte-colony stimulating factor(G-CSF); DA, daunorubicin and cytarabine; MA, mitoxantrone and cytarabine; NR, no response; CR, complete response; HDA, homoharringtonine, daunorubicin and cytarabine; D+ID-AC, daunorubicin and intermediate dose cytarabine; M+ID-AC, homoharringtonine and intermediate dose cytarabine; HA, homoharringtonine and cytarabine; HAA, homoharringtonine, aclamycin, and cytarabine; IA, idarubicin and cytarabine.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Su, Z.; Liu, X. Comment on Geoffroy, M.-C.; de Thé, H. Classic and Variants APLs, as Viewed from a Therapy Response. Cancers 2020, 12, 967. Cancers 2021, 13, 5883. https://doi.org/10.3390/cancers13235883

AMA Style

Su Z, Liu X. Comment on Geoffroy, M.-C.; de Thé, H. Classic and Variants APLs, as Viewed from a Therapy Response. Cancers 2020, 12, 967. Cancers. 2021; 13(23):5883. https://doi.org/10.3390/cancers13235883

Chicago/Turabian Style

Su, Zhan, and Xin Liu. 2021. "Comment on Geoffroy, M.-C.; de Thé, H. Classic and Variants APLs, as Viewed from a Therapy Response. Cancers 2020, 12, 967" Cancers 13, no. 23: 5883. https://doi.org/10.3390/cancers13235883

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop