1
|
Qin X, Chen X, Wang F, Zhong F, Zeng Y, Liu W. Huaier inhibits autophagy and promotes apoptosis in T-cell acute lymphoblastic leukemia by down-regulating SIRT1. Heliyon 2024; 10:e37313. [PMID: 39286166 PMCID: PMC11402646 DOI: 10.1016/j.heliyon.2024.e37313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
Objective Due to the high drug resistance and relapse rate of T-cell acute lymphoblastic leukemia (T-ALL), the prognosis is usually poor. Therefore, there is an urgent need to find safer and more effective therapeutic drugs. Huaier and its preparations, as adjuvant drugs, have been widely used in the treatment of solid tumors and other diseases. However, the application of Huaier in leukemia is rarely reported. In this study, we investigated the anti-tumor effect of Huaier on T- ALL and its underlying mechanism. Methods Jurkat and MOLT-4 cells were treated with Huaier. Cell viability was evaluated by CCK-8 assay. The morphological changes of apoptotic cells were observed by Hoechst 33258 staining. Cell apoptosis was analyzed by flow cytometry. The expression levels of related proteins were assessed by Western blot. Results The results showed that Huaier significantly inhibited the proliferation of Jurkat and MOLT-4 cells in a dose- and time-dependent manner, with IC50 of 2.37 ± 0.10 and 1.93 ± 0.07 mg/mL at 48 h, respectively. Morphological changes and increased number of apoptotic cells were observed by Hoechst 33258 staining and flow cytometry. The apoptosis rates of Jurkat and MOLT-4 cells in 4 mg/mL group were 50.67 ± 1.36 % and 49.97 ± 5.43 %, respectively. Huaier promoted the expression of Cytochrome c, Cleaved Caspase-3, Cleaved PARP, p53, LC3-Ⅱ and p62 proteins, while inhibited the expression of SIRT1, ATG7 and Beclin 1 proteins. Treatment with SRT1720 (SIRT1 agonist) combined with Huaier rescued Huaier-induced apoptosis and increased the expression of autophagy-related proteins. Conclusion Huaier inhibits autophagy and promotes apoptosis of T-ALL cells by down-regulating SIRT1, which may be a potential drug for the treatment of T-ALL.
Collapse
Affiliation(s)
- Xiang Qin
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| | - Xi Chen
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| | - Fan Wang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Fangfang Zhong
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| | - Yan Zeng
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| | - Wenjun Liu
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, 646000, China
| |
Collapse
|
2
|
Zhao J, Kang M, Li H, Rong L, Wang Y, Xue Y, Yao Y, Fang Y. QRICH1 suppresses pediatric T-cell acute lymphoblastic leukemia by inhibiting GRP78. Cell Death Dis 2024; 15:646. [PMID: 39227586 PMCID: PMC11371816 DOI: 10.1038/s41419-024-07040-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that commonly affects children and adolescents with a poor prognosis. The terminal unfolded protein response (UPR) is an emerging anti-cancer approach, although its role in pediatric T-ALL remains unclear. In our pediatric T-ALL cohort from different centers, a lower QRICH1 expression was found associated with a worse prognosis of pediatric T-ALL. Overexpression of QRICH1 significantly inhibited cell proliferation and stimulated apoptosis of T-ALL both in vitro and in vivo. Upregulation of QRICH1 significantly downregulated 78 KDa glucose-regulated protein (GRP78) and upregulated CHOP, thus activating the terminal UPR. Co-overexpression of GRP78 in T-ALL cells overexpressing QRICH1 partially reverted the inhibited proliferation and stimulated apoptosis. QRICH1 bound to the residues Asp212 and Glu155 of the nucleotide-binding domain (NBD) of GRP78, thereby inhibiting its ATP hydrolysis activity. In addition, QRICH1 was associated with endoplasmic reticulum (ER) stress in T-ALL, and overexpression of QRICH1 reversed drug resistance. Overall, low QRICH1 expression is an independent risk factor for a poor prognosis of pediatric T-ALL. By inhibiting GRP78, QRICH1 suppresses pediatric T-ALL.
Collapse
Affiliation(s)
- Ji'ou Zhao
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Meiyun Kang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Huimin Li
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Liucheng Rong
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yaping Wang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yao Xue
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yuqian Yao
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yongjun Fang
- Department of Hematology and Oncology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
3
|
Balducci E, Simonin M, Duployez N, Steimlé T, Dourthe ME, Villarese P, Ducassou S, Arnoux I, Cayuela JM, Balsat M, Courtois L, Andrieu G, Touzart A, Huguet F, Petit A, Ifrah N, Dombret H, Baruchel A, Macintyre E, Preudhomme C, Boissel N, Asnafi V. Genomic imbalance analysis provides new insight into prognostic factors in adult and pediatric T-ALL. Blood 2024; 144:988-1000. [PMID: 38518104 DOI: 10.1182/blood.2023022154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/24/2024] Open
Abstract
ABSTRACT Given the poor outcome of refractory and relapsing T-cell acute lymphoblastic leukemia (T-ALL), identifying prognostic markers is still challenging. Using single nucleotide polymorphism (SNP) array analysis, we provide a comprehensive analysis of genomic imbalances in a cohort of 317 newly diagnosed patients with T-ALL including 135 children and 182 adults with respect to clinical and biological features and outcomes. SNP array results identified at least 1 somatic genomic imbalance in virtually all patients with T-ALL (∼96%). Del(9)(p21) (∼70%) and UPD(9)p21)/CDKN2A/B (∼28%) were the most frequent genomic imbalances. Unexpectedly del(13)(q14)/RB1/DLEU1 (∼14%) was the second most frequent copy number variant followed by del(6)(q15)/CASP8AP2 (∼11%), del(1)(p33)/SIL-TAL1 (∼11%), del(12)(p13)ETV6/CDKN1B (∼9%), del(18)(p11)/PTPN2 (∼9%), del(1)(p36)/RPL22 (∼9%), and del(17)(q11)/NF1/SUZ12 (∼8%). SNP array also revealed distinct profiles of genomic imbalances according to age, immunophenotype, and oncogenetic subgroups. In particular, adult patients with T-ALL demonstrated a significantly higher incidence of del(1)(p36)/RPL22, and del(13)(q14)/RB1/DLEU1, and lower incidence of del(9)(p21) and UPD(9p21)/CDKN2A/B. We determined a threshold of 15 genomic imbalances to stratify patients into high- and low-risk groups of relapse. Survival analysis also revealed the poor outcome, despite the low number of affected cases, conferred by the presence of chromothripsis (n = 6, ∼2%), del(16)(p13)/CREBBP (n = 15, ∼5%) as well as the newly-identified recurrent gain at 6q27 involving MLLT4 (n = 10, ∼3%). Genomic complexity, del(16)(p13)/CREBBP and gain at 6q27 involving MLLT4, maintained their significance in multivariate analysis for survival outcome. Our study thus demonstrated that whole genome analysis of imbalances provides new insights to refine risk stratification in T-ALL. This trial was registered at www.ClinicalTrials.gov as #NCT00222027 and #NCT00327678, and as #FRALLE 2000T trial.
Collapse
Affiliation(s)
- Estelle Balducci
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Mathieu Simonin
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Nicolas Duployez
- Laboratory of hematology, Biology and Pathology Center, CHU Lille, Lille, France
- INSERM U1277 CANTHER, University Lille, Lille, France
| | - Thomas Steimlé
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Marie-Emilie Dourthe
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Patrick Villarese
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Stéphane Ducassou
- Department of Pediatric Hematology-Oncology, Bordeaux University Hospital, Bordeaux, France
| | - Isabelle Arnoux
- Hematology Laboratory, Marseille University Hospital Timone, Marseille, France
| | - Jean-Michel Cayuela
- Laboratory of Hematology and EA 3518 University Hospital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Université de Paris, Paris, France
| | - Marie Balsat
- Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Lyon, France
| | - Lucien Courtois
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Guillaume Andrieu
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Aurore Touzart
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Françoise Huguet
- Hematology Department, Institut Universitaire du Cancer-Oncopole, CHU de Toulouse, Toulouse, France
| | - Arnaud Petit
- Department of Pediatric Hematology and Oncology, Assistance Publique-Hôpitaux de Paris (AP-HP), GH HUEP, Armand Trousseau Hospital, Paris, France
| | - Norbert Ifrah
- PRES LUNAM, CHU Angers Service des Maladies du Sang et INSERM U 892, Angers, France
| | - Hervé Dombret
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - André Baruchel
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
- Department of Pediatric Hematology and Immunology, University Hospital Robert Debré, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Elizabeth Macintyre
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| | - Claude Preudhomme
- Laboratory of hematology, Biology and Pathology Center, CHU Lille, Lille, France
- INSERM U1277 CANTHER, University Lille, Lille, France
| | - Nicolas Boissel
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - Vahid Asnafi
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Paris, France
| |
Collapse
|
4
|
Kroeze E, Kleisman MM, Kester LA, Scheijde‐Vermeulen MA, Sonneveld E, Buijs‐Gladdines JGC, Hagleitner MM, Meyer‐Wentrup FAG, Veening MA, Beishuizen A, Meijerink JPP, Loeffen JLC, Kuiper RP. NOTCH1 fusions in pediatric T-cell lymphoblastic lymphoma: A high-risk subgroup with CCL17 (TARC) levels as diagnostic biomarker. Hemasphere 2024; 8:e117. [PMID: 38948925 PMCID: PMC11208779 DOI: 10.1002/hem3.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 07/02/2024] Open
Abstract
Twenty percent of children with T-cell lymphoblastic lymphoma (T-LBL) will relapse and have an extremely poor outcome. Currently, we can identify a genetically low-risk subgroup in pediatric T-LBL, yet these high-risk patients who need intensified or alternative treatment options remain undetected. Therefore, there is an urgent need to recognize these high-risk T-LBL patients through identification of molecular characteristics and biomarkers. By using RNA sequencing which was performed in 29/49 T-LBL patients who were diagnosed in the Princess Maxima Center for Pediatric Oncology between 2018 and 2023, we discovered a previously unknown high-risk biological subgroup of children with T-LBL. This subgroup is characterized by NOTCH1 gene fusions, found in 21% of our T-LBL cohort (6/29). All patients presented with a large mediastinal mass, pleural/pericardial effusions, and absence of blasts in the bone marrow, blood, and central nervous system. Blood CCL17 (C-C Motif Chemokine Ligand 17, TARC) levels were measured at diagnosis in 26/29 patients, and all six patients with NOTCH1 gene fusions patients exclusively expressed highly elevated blood CCL17 levels, defining a novel and previously not known clinically relevant biomarker for T-cell lymphoblastic lymphoma. Four out of these six patients relapsed during therapy, a fifth developed a therapy-related acute myeloid leukemia during maintenance therapy. These data indicate that T-LBL patients with a NOTCH1 fusion have a high risk of relapse which can be easily identified using a blood CCL17 screening at diagnosis. Further molecular characterization through NOTCH1 gene fusion analysis offers these patients the opportunity for treatment intensification or new treatment strategies.
Collapse
Affiliation(s)
- Emma Kroeze
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | | - Edwin Sonneveld
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | | | | - Auke Beishuizen
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Jules P. P. Meijerink
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Present address:
Acerta‐Pharma (AstraZeneca)OssThe Netherlands
| | | | - Roland P. Kuiper
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
- Department of GeneticsUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
5
|
Sun H, Zhu Y, Li J, Zhao L, Yang G, Yan Z, Zhang S. PICALM::MLLT10 may indicate a new subgroup of acute leukemias with miscellaneous immunophenotype and poor initial treatment response but showing sensitivity to venetoclax. EJHAEM 2024; 5:565-572. [PMID: 38895061 PMCID: PMC11182389 DOI: 10.1002/jha2.922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Abstract
The PICALM::MLLT10 fusion gene is a rare but recurrent event in acute leukemia (AL) associated with poor prognosis. It is still confused whether PICALM::MLLT10 can solely correspond to acute myeloid leukemia (AML) or acute lymphoblastic leukemia (ALL) or acute leukemias of ambiguous lineage (ALAL). Here, we reported a series of PICALM::MLLT10 positive AL patients with miscellaneous immunophenotype including T-ALL, ALAL, AML, and B-ALL, complex karyotype, half of extramedullary disease (EMD), frequently concomitant PHF6 mutation, and poor initial treatment response to standard chemotherapy aiming to different immunophenotype, but showing sensitivity to combining chemotherapy especially integrated with venetoclax, suggesting this fusion gene may indicate a new subgroup of AL. Eighteen PICALM::MLLT10 positive patients of 533 AL patients (18/533, 3.4%) were identified by RNA sequencing in our center. We found PICALM::MLLT10 positive AL showing miscellaneous immunophenotype, higher expression of leukemic stemness genes and lower expression of biomarkers of venetoclax resistance, more extramedullary involvement, and especially poor response to conventional induction chemotherapy, but may benefit from venetoclax as well as low-dose Ara-C, granulocyte colony-stimulating factor (G-CSF), and anthracyclines combination chemotherapy. Sequential hematopoietic stem cell transplantation (HSCT) after chemotherapy combined with venetoclax may further improve long-term survival in AL patients with complete remission (CR) even measurable residual disease (MRD) positive.
Collapse
Affiliation(s)
- Haimin Sun
- Department of Hematology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yongmei Zhu
- Shanghai Institute of HematologyState Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jianfeng Li
- Shanghai Institute of HematologyState Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lingling Zhao
- Shanghai Institute of HematologyState Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Guang Yang
- Shanghai Institute of HematologyState Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zeying Yan
- Department of Hematology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sujiang Zhang
- Department of Hematology, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai Institute of HematologyState Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
6
|
Nguyen PT, Shimojukkoku Y, Kajiya Y, Oku Y, Tomishima A, Shima K, Sasahira T. Gene alterations in the nuclear transport receptor superfamily: A study of head and neck cancer. PLoS One 2024; 19:e0300446. [PMID: 38820302 PMCID: PMC11142601 DOI: 10.1371/journal.pone.0300446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 02/28/2024] [Indexed: 06/02/2024] Open
Abstract
In cancer cells, the nuclear transport system is often disrupted, leading to abnormal localization of nuclear proteins and altered gene expression. This disruption can arise from various mechanisms such as mutations in genes that regulate nuclear transport, altered expression of transport proteins, and changes in nuclear envelope structure. Oncogenic protein build-up in the nucleus due to the disturbance in nuclear transport can also boost tumor growth and cell proliferation. In this study, we performed bioinformatic analyses of 23 key nuclear transport receptors using genomic and transcriptomic data from pancancer and head and neck squamous cell carcinoma (HNSCC) datasets from The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia and found that the total alteration frequency of 23 nuclear transport receptors in 2691 samples of the PCAWG Consortium was 42.1% and a high levels of genetic alterations was significantly associated with poor overall survival. Amplification was the most common type of genetic alterations, and results in the overexpression of nuclear transport receptors in HNSCC compared to normal tissues. Furthermore, our study revealed that seven out of eight cell cycle genes (CDK1, CDK2, CDK4, CDK6, CCNA1, CCNB1, and CCNE2) were significantly and positively correlated with nuclear transport receptor genes in TCGA pancancer and CCLE datasets. Additionally, functional enrichment analysis showed that nuclear transport receptor genes were mainly enriched in the adhesion junction, cell cycle, ERBB, MAPK, MTOR and WNT signaling pathways.
Collapse
Affiliation(s)
- Phuong Thao Nguyen
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| | - Yudai Shimojukkoku
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| | - Yuka Kajiya
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| | - Yasunobu Oku
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| | - Ayami Tomishima
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| | - Kaori Shima
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| | - Tomonori Sasahira
- Department of Molecular Oral Pathology and Oncology, Graduate School of Medical and Dental Science, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
7
|
van Outersterp I, Tasian SK, Reichert CEJ, Boeree A, de Groot-Kruseman HA, Escherich G, Boer JM, den Boer ML. Tyrosine kinase inhibitor response of ABL-class acute lymphoblastic leukemia: the role of kinase type and SH3 domain. Blood 2024; 143:2178-2189. [PMID: 38394665 PMCID: PMC11143520 DOI: 10.1182/blood.2023023120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/10/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
ABSTRACT Acute lymphoblastic leukemia (ALL) with fusions of ABL-class tyrosine kinase genes other than BCR::ABL1 occurs in ∼3% of children with ALL. The tyrosine kinase genes involved in this BCR::ABL1-like (Ph-like) subtype include ABL1, PDGFRB, ABL2, and CSF1R, each of which has up to 10 described partner genes. ABL-class ALL resembles BCR::ABL1-positive ALL with a similar gene expression profile, poor response to chemotherapy, and sensitivity to tyrosine kinase inhibitors (TKIs). There is a lack of comprehensive data regarding TKI sensitivity in the heterogeneous group of ABL-class ALL. We observed variability in TKI sensitivity within and among each ABL-class tyrosine kinase gene subgroup. We showed that ALL samples with fusions for any of the 4 tyrosine kinase genes were relatively sensitive to imatinib. In contrast, the PDGFRB-fused ALL samples were less sensitive to dasatinib and bosutinib. Variation in ex vivo TKI response within the subset of samples with the same ABL-class tyrosine kinase gene was not associated with the ALL immunophenotype, 5' fusion partner, presence or absence of Src-homology-2/3 domains, or deletions of IKZF1, PAX5, or CDKN2A/B. In conclusion, the tyrosine kinase gene involved in ABL-class ALL is the main determinant of TKI sensitivity and relevant for specific TKI selection.
Collapse
Affiliation(s)
| | - Sarah K. Tasian
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| | | | - Aurélie Boeree
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Monique L. den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology and Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Xiao M, Zhou J, Zhu X, He Y, Wang F, Zhang Y, Mo X, Han W, Wang J, Wang Y, Chen H, Chen Y, Zhao X, Chang Y, Xu L, Liu K, Huang X, Zhang X. A prognostic score system in adult T-cell acute lymphoblastic leukemia after hematopoietic stem cell transplantation. Bone Marrow Transplant 2024; 59:496-504. [PMID: 38267585 DOI: 10.1038/s41409-024-02211-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
Adult T-cell acute lymphoblastic leukemia (T-ALL) is highly aggressive with poor prognoses, while hematopoietic stem cell transplantation (HSCT) is a curable option. However, no transplant-specific prognostic model for adult T-ALL is available. We identified 301 adult T-ALL patients who received HSCT at our hospital between 2010 and 2022. These patients were randomly assigned at a 7:3 ratio to a derivation group of 210 patients and a validation group of 91 patients. Next, we developed a prognostic risk score system for adult T-ALL with HSCT, which we named COMM, including 4 predictors (central nervous system involvement, Non-CR1 (CR2+ or NR) at HSCT, minimal residual disease (MRD) ≥ 0.01% after first induction therapy, and MRD ≥ 0.01% before HSCT). Patients were categorized into three risk groups, low-risk (0), intermediate-risk (1-4), and high-risk (5-12), and their 3-year overall survival (OS) were 87.5% (95%CI, 78-93%), 65.7% (95%CI, 53-76%) and 20% (95%CI, 10-20%; P < 0.001), respectively. The area under the subject operating characteristic curve for 2-, 3- or 5-year OS in the derivation cohort and in the validation cohort were all greater than 0.75. Based on internal validation, COMM score system proved to be a reliable prognostic model that could discriminate and calibrate well. We expect that the first prognostic model in adults T-ALL after HSCT can provide a reference of prognostic consultation for patients and families, and also contribute to future research to develop risk adapted interventions for high-risk populations.
Collapse
Affiliation(s)
- Mengyu Xiao
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Jianying Zhou
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaolu Zhu
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yun He
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Fengrong Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yuanyuan Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaodong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Wei Han
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Jingzhi Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Huan Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yuhong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiangyu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Lanping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Kaiyan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Collaborative Innovation Center of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.
| |
Collapse
|
9
|
Mark C, Meshinchi S, Joyce B, Gibson B, Harrison C, Bergmann AK, Goemans BF, Pronk CJH, Lapillonne H, Leverger G, Antoniou E, Schneider M, Attarbaschi A, Dworzak M, Stary J, Tomizawa D, Ebert S, Lejman M, Kolb EA, Schmiegelow K, Hasle H, Abla O. Treatment outcomes of childhood PICALM::MLLT10 acute leukaemias. Br J Haematol 2024; 204:576-584. [PMID: 37743097 DOI: 10.1111/bjh.19067] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/18/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
The prognostic impact of PICALM::MLLT10 status in childhood leukaemia is not well described. Ten International Berlin Frankfurt Münster-affiliated study groups and the Children's Oncology Group collaborated in this multicentre retrospective study. The presence of the PICALM::MLLT10 fusion gene was confirmed by fluorescence in situ hybridization and/or RNA sequencing at participating sites. Ninety-eight children met the study criteria. T-cell acute lymphoblastic leukaemia (T-ALL) and acute myeloid leukaemia (AML) predominated 55 (56%) and 39 (40%) patients, respectively. Most patients received a chemotherapy regimen per their disease phenotype: 58% received an ALL regimen, 40% an AML regimen and 1% a hybrid regimen. Outcomes for children with PICALM::MLLT10 ALL were reasonable: 5-year event-free survival (EFS) 67% and 5-year overall survival (OS) 76%, but children with PICALM::MLLT10 AML had poor outcomes: 5-year EFS 22% and 5-year OS 26%. Haematopoietic stem cell transplant (HSCT) did not result in a significant improvement in outcomes for PICALM::MLLT10 AML: 5-year EFS 20% for those who received HSCT versus 23% for those who did not (p = 0.6) and 5-year OS 37% versus 36% (p = 0.7). In summary, this study confirms that PICALM::MLLT10 AML is associated with a dismal prognosis and patients cannot be salvaged with HSCT; exploration of novel therapeutic options is warranted.
Collapse
Affiliation(s)
- Catherine Mark
- Division of Hematology/Oncology, Toronto Hospital for Sick Children, Toronto, Ontario, Canada
| | - Soheil Meshinchi
- Fred Hutchinson Cancer Research Centre, Seattle, Washington, USA
| | - Brooklyn Joyce
- Division of Hematology/Oncology, Toronto Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brenda Gibson
- Royal Hospital for Sick Children, Glasgow, Scotland, UK
| | | | | | - Bianca F Goemans
- Princess Maxima Centre for Pediatric Oncology, Utrecht, The Netherlands
| | | | | | - Guy Leverger
- Hôpital d'enfants Armand Trousseau, Paris, France
| | | | | | - Andishe Attarbaschi
- Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Michael Dworzak
- Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Jan Stary
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Daisuke Tomizawa
- Division of Leukemia and Lymphoma, Children's Cancer Centre, National Centre for Child Health and Development, Tokyo, Japan
| | - Sabine Ebert
- Clinic of Pediatric Hematology and Oncology, University Medical Centre, Hamburg, Germany
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, Lublin, Poland
| | - E Anders Kolb
- Nemours Children's Hospital, Wilmington, Delaware, USA
| | | | - Henrik Hasle
- Hematology/Oncology, Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| | - Oussama Abla
- Division of Hematology/Oncology, Toronto Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
10
|
De Bie J, Quessada J, Tueur G, Lefebvre C, Luquet I, Toujani S, Cuccuini W, Lafage-Pochitaloff M, Michaux L. Cytogenetics in the management of T-cell acute lymphoblastic leukemia (T-ALL): Guidelines from the Groupe Francophone de Cytogénétique Hématologique (GFCH). Curr Res Transl Med 2023; 71:103431. [PMID: 38016418 DOI: 10.1016/j.retram.2023.103431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023]
Abstract
Molecular analysis is the hallmark of T-cell acute lymphoblastic leukemia (T-ALL) categorization. Several T-ALL sub-groups are well recognized based on the aberrant expression of specific transcription factors. This recently resulted in the implementation of eight provisional T-ALL entities into the novel 2022 International Consensus Classification, albeit not into the updated World Health Organization classification system. Despite this extensive molecular characterization, cytogenetic analysis remains the backbone of T-ALL diagnosis in many countries as chromosome banding analysis and fluorescence in situ hybridization are relatively inexpensive techniques to obtain results of diagnostic, prognostic and therapeutic interest. Here, we provide an overview of recurrent chromosomal abnormalities detectable in T-ALL patients and propose guidelines regarding their detection. By referring in parallel to the more general molecular classification approach, we hope to offer a diagnostic framework useful in a broad clinical genetic setting.
Collapse
Affiliation(s)
- Jolien De Bie
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium
| | - Julie Quessada
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France; CRCM, Inserm UMR1068, CNRS UMR7258, Aix Marseille Université U105, Institut Paoli Calmettes, Marseille 13009, France
| | - Giulia Tueur
- Laboratoire d'hématologie, Hôpital Avicenne, AP-HP, Bobigny 93000, France
| | - Christine Lefebvre
- Unité de Génétique des Hémopathies, Service d'Hématologie Biologique, CHU Grenoble Alpes, Grenoble 38000, France
| | - Isabelle Luquet
- Laboratoire d'Hématologie, CHU Toulouse (IUCT-O), Toulouse 31000, France
| | - Saloua Toujani
- Service de Cytogénétique et Biologie Cellulaire, CHU de Rennes, Rennes 35033, France
| | - Wendy Cuccuini
- Laboratoire d'Hématologie, Unité de Cytogénétique, Hôpital Saint-Louis, AP-HP, Paris 75010, France
| | - Marina Lafage-Pochitaloff
- Laboratoire de Cytogénétique Hématologique, Département d'Hématologie, CHU Timone, APHM, Aix Marseille Université, Marseille 13005, France
| | - Lucienne Michaux
- Center for Human Genetics, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium; Katholieke Universiteit Leuven, Leuven 3000, Belgium.
| |
Collapse
|
11
|
van Outersterp I, van der Velden VH, Hoogeveen PG, Vaitkevičienė GE, Sonneveld E, van Haaften G, Kuiper RP, zur Stadt U, Escherich G, Boer JM, den Boer ML. ABL-class Genomic Breakpoint Q-PCR: A Patient-specific Approach for MRD Monitoring in Acute Lymphoblastic Leukemia. Hemasphere 2023; 7:e967. [PMID: 37736661 PMCID: PMC10511034 DOI: 10.1097/hs9.0000000000000967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023] Open
Affiliation(s)
| | - Vincent H.J. van der Velden
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Patricia G. Hoogeveen
- Department of Immunology, Laboratory Medical Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Goda E. Vaitkevičienė
- Faculty of Medicine, Vilnius University, Lithuania
- Center for Pediatric Oncology and Hematology, Vilnius University Hospital Santaros Klinikos, Lithuania
| | - Edwin Sonneveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Dutch Childhood Oncology Group, Utrecht, The Netherlands
| | - Gijs van Haaften
- Department of Genetics, University Medical Center Utrecht, The Netherlands
| | - Roland P. Kuiper
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Genetics, University Medical Center Utrecht, The Netherlands
| | - Udo zur Stadt
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Gabriele Escherich
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg Eppendorf, Germany
| | - Judith M. Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Monique L. den Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Pediatric Oncology and Hematology, Erasmus MC - Sophia Children’s Hospital, Rotterdam, The Netherlands
| |
Collapse
|
12
|
Li W, Hu JK, Hu MG. CDK6: an attractive therapeutic target for T-ALL/LBL. Expert Opin Ther Targets 2023; 27:1087-1096. [PMID: 37975616 DOI: 10.1080/14728222.2023.2285775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 11/16/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Human T-cell acute lymphoblastic leukemia/T-cell lymphoblastic lymphoma (T-ALL/LBL) is a type of cancer that originates from the bone marrow and spreads quickly to other organs. Long-term survival rate with current available chemotherapy is less than 20%. Despite the potentially huge market, a truly effective and safe therapy for T-ALL/LBL is elusive. Thus, it is imperative to identify new therapeutic ways to target essential pathways in T-ALL that regulate the proliferation and survival of these cancer cells. AREAS COVERED The role of the Cyclin-dependent kinase 6 (CDK6) pathway in human T-ALL is of significant interest with major clinical/translational relevance. This review covers the recent advances in elucidating the essential roles of CDK6 and its closely regulated networks in proliferation, survival, and metabolism of T-ALL cells, with new insight into its mechanisms of action which hopefully could trigger the identification of new therapeutic avenues. EXPERT OPINION Animal models showed that inhibition of CDK6 and its related networks blocked initiation, growth, and survival of T-ALL in vivo. Numerous clinical trials of CDK4/6 inhibitors are ongoing in T-ALL. Specific CDK6 inhibitors alone or novel combination regimens may hopefully delay the progression, or even reverse the symptoms of T-ALL, leading to disease eradication and cure.
Collapse
Affiliation(s)
- Wei Li
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, USA
| | - Jamie Katy Hu
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Miaofen G Hu
- Department of Medicine, Division of Hematology and Oncology, Tufts Medical Center, Boston, USA
| |
Collapse
|
13
|
Soler G, Ouedraogo ZG, Goumy C, Lebecque B, Aspas Requena G, Ravinet A, Kanold J, Véronèse L, Tchirkov A. Optical Genome Mapping in Routine Cytogenetic Diagnosis of Acute Leukemia. Cancers (Basel) 2023; 15:cancers15072131. [PMID: 37046792 PMCID: PMC10093111 DOI: 10.3390/cancers15072131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/14/2023] Open
Abstract
Cytogenetic aberrations are found in 65% of adults and 75% of children with acute leukemia. Specific aberrations are used as markers for the prognostic stratification of patients. The current standard cytogenetic procedure for acute leukemias is karyotyping in combination with FISH and RT-PCR. Optical genome mapping (OGM) is a new technology providing a precise identification of chromosomal abnormalities in a single approach. In our prospective study, the results obtained using OGM and standard techniques were compared in 29 cases of acute myeloid (AML) or lymphoblastic leukemia (ALL). OGM detected 73% (53/73) of abnormalities identified by standard methods. In AML cases, two single clones and three subclones were missed by OGM, but the assignment of patients to cytogenetic risk groups was concordant in all patients. OGM identified additional abnormalities in six cases, including one cryptic structural variant of clinical interest and two subclones. In B-ALL cases, OGM correctly detected all relevant aberrations and revealed additional potentially targetable alterations. In T-ALL cases, OGM characterized a complex karyotype in one case and identified additional abnormalities in two others. In conclusion, OGM is an attractive alternative to current multiple cytogenetic testing in acute leukemia that simplifies the procedure and reduces costs.
Collapse
Affiliation(s)
- Gwendoline Soler
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, 63000 Clermont-Ferrand, France
| | - Zangbéwendé Guy Ouedraogo
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, 63000 Clermont-Ferrand, France
- Service de Biochimie et Génétique Moléculaire, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
- CNRS, INSERM, iGReD, Université Clermont Auvergne, 63001 Clermont-Ferrand, France
| | - Carole Goumy
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, 63000 Clermont-Ferrand, France
- INSERM U1240 Imagerie Moléculaire et Stratégies Théranostiques, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | | | - Gaspar Aspas Requena
- Hématologie Clinique Adulte et de Thérapie Cellulaire, CHU Estaing, 63100 Clermont-Ferrand, France
| | - Aurélie Ravinet
- Hématologie Clinique Adulte et de Thérapie Cellulaire, CHU Estaing, 63100 Clermont-Ferrand, France
| | - Justyna Kanold
- Service d'Hématologie et d'Oncologie Pédiatrique et Unité CRECHE (Centre de REcherche Clinique CHez l'Enfant), CHU Estaing, 63100 Clermont-Ferrand, France
| | - Lauren Véronèse
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, 63000 Clermont-Ferrand, France
- Clonal Heterogeneity and Leukemic Environment in Therapy Resistance of Chronic Leukemias (CHELTER), EA7453, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| | - Andrei Tchirkov
- Cytogénétique Médicale, CHU Clermont-Ferrand, CHU Estaing, 63000 Clermont-Ferrand, France
- Clonal Heterogeneity and Leukemic Environment in Therapy Resistance of Chronic Leukemias (CHELTER), EA7453, Université Clermont Auvergne, 63000 Clermont-Ferrand, France
| |
Collapse
|
14
|
Chen Y, Wang Q, Cen J, Xu C, Tao TT, Xie J, Shen W, Gong Y, Pan J, Yao L. Blast phase of chronic myeloid leukemia with concurrent BCR::ABL1 and SET::NUP214: A report of two cases. Mol Carcinog 2023; 62:117-121. [PMID: 36321418 DOI: 10.1002/mc.23480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/21/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm defined by the presence of t(9;22)(q34;q11.2)/BCR::ABL1. Additional chromosomal abnormalities play an important role in the progression to CML. However, the additional fusion gene was rarely reported such as CBFB::MYH11. In this report, we described two cases of the co-occurrence of BCR::ABL1 and SET::NUP214 in CML-BP for the first time, which is associated with poor outcomes during tyrosine kinase inhibitor (TKI) treatment. Meanwhile, we retrospectively analyzed SET::NUP214 fusion transcript of the two cases at initial diagnosis of the CML chronic phase by quantitative RT-PCR, and detected at a ratio of 1.63% and 1.50%, respectively. SET::NUP214 may promote disease progression during the transformation of CML. This study highlights the importance of extended molecular testing at the initial diagnosis of CML-CP at TKI resistance and/or disease transformation.
Collapse
Affiliation(s)
- Yan Chen
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian Wang
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiannong Cen
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chao Xu
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ting-Ting Tao
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jundan Xie
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenhong Shen
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanlei Gong
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinlan Pan
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Yao
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
15
|
Kumari S, Ali MS, Singh J, Arora M, Verma D, Pandey AK, Benjamin M, Bakhshi S, Palanichamy JK, Sharma A, Singh I, Tanwar P, Singh AR, Pushpam D, Qamar I, Chopra A. Prognostic utility of key copy number alterations in T cell acute lymphoblastic leukemia. Hematol Oncol 2022; 40:577-587. [DOI: 10.1002/hon.3030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/26/2022] [Accepted: 05/21/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Sarita Kumari
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
- School of Biotechnology Gautam Buddha University Uttar Pradesh201312 India
| | - Md Shadab Ali
- Department of Pulmonary Medicine and Sleep Disorders All India Institute of Medical Sciences New Delhi New Delhi110029 India
| | - Jay Singh
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Mohit Arora
- Department of Biochemistry All India Institute of Medical Sciences New Delhi110029 India
| | - Deepak Verma
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Avanish Kumar Pandey
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Mercilena Benjamin
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Sameer Bakhshi
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | | | - Atul Sharma
- Department of Medical Oncology Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Inder Singh
- Department of Neurology All India Institute of Medical Sciences New Delhi New Delhi110029 India
| | - Pranay Tanwar
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Amar Ranjan Singh
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Deepam Pushpam
- Department of Medical Oncology Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| | - Imteyaz Qamar
- School of Biotechnology Gautam Buddha University Uttar Pradesh201312 India
| | - Anita Chopra
- Laboratory Oncology Unit Dr. BRA‐IRCH All India Institute of Medical Sciences New Delhi110029 India
| |
Collapse
|
16
|
Early T-Cell Precursor ALL and Beyond: Immature and Ambiguous Lineage T-ALL Subsets. Cancers (Basel) 2022; 14:cancers14081873. [PMID: 35454781 PMCID: PMC9030030 DOI: 10.3390/cancers14081873] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immature T-cell acute lymphoblastic leukemias englobes a wide range of low prevalence subtypes, not well identified, that in some cases overlap with myeloid lineage subtypes. Globally, this “grey zone” of immature leukemias, are difficult to precisely diagnose using a classical immunophenotypic approach. Interesting, genomic data collected during last years has shown that these subtypes share several genomic alterations, raising the question of how their phenotypes reflect distinct AL entities. Here we provide a systematic overview of the genetic events associated with immature T-ALL and outline their relationship with treatment choices and outcomes. Our goal is to offer a basis for using the genetic information for new diagnostic algorithms. An immunogenetic classification of these immature subtypes will better stratify patients and improve their management with more efficient and personalized therapeutic options. Abstract A wide range of immature acute leukemias (AL), ranging from acute myeloid leukemias with minimal differentiation to acute leukemias with an ambiguous lineage, i.e., acute undifferentiated leukemias and mixed phenotype acute leukemia with T- or B-plus myeloid markers, cannot be definitely assigned to a single cell lineage. This somewhat “grey zone” of AL expresses partly overlapping features with the most immature forms of T-cell acute lymphoblastic leukemia (T-ALL), i.e., early T-cell precursor ALL (ETP-ALL), near-ETP-ALL, and pro-T ALL. These are troublesome cases in terms of precise diagnosis because of their similarities and overlapping phenotypic features. Moreover, it has become evident that they share several genomic alterations, raising the question of how their phenotypes reflect distinct AL entities. The aim of this review was to provide a systematic overview of the genetic events associated with immature T-ALL and outline their relationship with treatment choices and outcomes, especially looking at the most recent preclinical and clinical studies. We wish to offer a basis for using the genetic information for new diagnostic algorithms, in order to better stratify patients and improve their management with more efficient and personalized therapeutic options. Understanding the genetic profile of this high-risk T-ALL subset is a prerequisite for changing the current clinical scenario.
Collapse
|