1
|
Huang SM, Chen HQ, Liu LT, Zhang YT, Wang J, Zhou DH, Fang JP, Xu LH. Clinical characteristics and prognostic analysis of CDKN2A/2B gene in pediatric acute lymphoblastic leukemia: a retrospective case-control study. Hematology 2025; 30:2439606. [PMID: 39676312 DOI: 10.1080/16078454.2024.2439606] [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: 08/07/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
In this retrospective case-control study involving 424 pediatric patients diagnosed with Pediatric Acute Lymphoblastic Leukemia (ALL), the investigation focused on analyzing the clinical characteristics and prognosis associated with the Cyclin-dependent kinase inhibitor 2A/2B (CDKN2A/2B) gene. Treatment and evaluation followed the South China Children's Leukemia Group-ALL-2016 protocol (SCCLG-ALL-2016). Among the cohort, 92 patients (21.7%) exhibited CDKN2A/2B gene deletions, with 11.1% homozygous and 10.6% heterozygous deletions. Notably, ALL patients that do have CDKN2A/2B gene deletions tended to present at an older age (P = 0.001), demonstrate hepatosplenomegaly on palpation (P < 0.001), and exhibit a higher incidence of Central nervous system leukemia (CNSL) (P = 0.037) and T-ALL (P = 0.007). A significant correlation was observed between ALL that does have CDKN2A/2B gene deletions and ETV6::RUNX1-positive (8.7% vs. 19.3%, P = 0.017) and IKZF1 gene deletions (20.7% vs. 8.4%, P = 0.001). Survival analysis of 392 patients revealed no significant differences in 5-year relapse, Overall survival (OS), or Event-free survival (EFS) between ALL that does/ does not have CDKN2A/2B gene deletions. Subgroup analysis highlighted poorer prognosis among hepatosplenomegaly patients in the CDKN2A/2B gene deletion group, with a 5-year EFS of 81.8%, 95%CI (0.695-0.963), P = 0.05. Hepatosplenomegaly emerged as the most significant prognostic factor for EFS [HR = 2.306, 95%CI (1.192-4.461), P = 0.013]. Cox regression analyses identified covariates influencing prognosis, ALL with the CDKN2A/2B gene showing no significant impact on outcomes. In conclusion, while ALL that does have CDKN2A/2B gene deletions is associated with certain clinical characteristics and genetic aberrations, they did not significantly impact OS or EFS. Furthermore, subgroup analysis revealed a potential prognostic role of ALL that does have CDKN2A/2B deletions presenting with hepatosplenomegaly on palpation, emphasizing the importance of comprehensive risk stratification in treatment decision-making for this subgroup.
Collapse
Affiliation(s)
- Shi-Mei Huang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hui-Qin Chen
- Department of Pediatrics, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li-Ting Liu
- Department of Pediatrics, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ya-Ting Zhang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian Wang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dun-Hua Zhou
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian-Pei Fang
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Lu-Hong Xu
- Children's Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
2
|
Shao X, Yokomori R, Ong JZL, Shen H, Kappei D, Chen L, Yeoh AEJ, Tan SH, Sanda T. Transcriptional regulatory program controlled by MYB in T-cell acute lymphoblastic leukemia. Leukemia 2024; 38:2573-2584. [PMID: 39488662 DOI: 10.1038/s41375-024-02455-9] [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: 06/18/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
The transcription factor MYB is frequently upregulated in T-cell acute lymphoblastic leukemia (T-ALL), a hematological malignancy originating from T-cell precursors. Here, we demonstrate that MYB plays a crucial role by regulating genes essential for T-ALL pathogenesis. Integrative analysis reveals a long MYB isoform, ENST00000367814.8, which is dominantly expressed and confers a proliferative advantage in T-ALL cells. Rapid depletion of MYB via dTAG-mediated protein degradation affects a large number of genes, which can be classified into early response or late response genes based on their kinetics. Early response genes include many genes involved in hematopoiesis, such as TAL1, RUNX1, GATA3, IKZF2, and CXCR4. Their expression can be recovered at later time-points, suggesting the presence of a negative feedback loop mechanism. In contrast, late response genes, which are continuously downregulated after MYB depletion, includes many genes involved in cell proliferation as well as TAL1 targets, thereby affecting the cellular phenotype.
Collapse
Affiliation(s)
- Xiaoman Shao
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Rui Yokomori
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Jolynn Zu Lin Ong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Haoqing Shen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Leilei Chen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Allen Eng Juh Yeoh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pediatrics, National University of Singapore, Singapore, Singapore
| | - Shi Hao Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| |
Collapse
|
3
|
Kaur P, Sharma P, Bhatia P, Singh M. Recent advances on biogenesis, functions and therapeutic potential of long noncoding RNAs in T cell acute lymphoblastic leukemia. Discov Oncol 2024; 15:729. [PMID: 39612075 DOI: 10.1007/s12672-024-01618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
T-cell Acute Lymphoblastic Leukemia (T-ALL) is a highly aggressive form of ALL with at least 25% relapse rates. The high relapse rates are often linked to poor prognoses. More detailed studies for novel therapeutic targets for the treatment of T-ALL are required as the genetic and transcriptomic data currently available on T-ALL pathophysiology is insufficient. Long non-coding RNAs are emerging as important players in the regulation of tumour proliferation and metastasis. Studies on various cancers have revealed their potential as biomarkers and therapeutic targets in treatment. This review describes the characterization, biosynthesis, and role of long non-coding RNA in T-ALL and highlights their potential as next generation molecule in development of promising diagnostic, prognostic and/or therapeutic markers.
Collapse
Affiliation(s)
- Parminder Kaur
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Sharma
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Prateek Bhatia
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Minu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
4
|
Rahman S, Bloye G, Farah N, Demeulemeester J, Costa JR, O'Connor D, Pocock R, Rapoz-D'Silva T, Turna A, Wang L, Lee S, Fielding AK, Roels J, Jaksik R, Dawidowska M, Van Vlierberghe P, Hadjur S, Hughes JR, Davies JOJ, Gutierrez A, Kelliher MA, Van Loo P, Dawson MA, Mansour MR. Focal deletions of a promoter tether activate the IRX3 oncogene in T-cell acute lymphoblastic leukemia. Blood 2024; 144:2319-2326. [PMID: 39316719 DOI: 10.1182/blood.2024024300] [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: 02/14/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
ABSTRACT Oncogenes can be activated in cis through multiple mechanisms including enhancer hijacking events and noncoding mutations that create enhancers or promoters de novo. These paradigms have helped parse somatic variation of noncoding cancer genomes, thereby providing a rationale to identify noncanonical mechanisms of gene activation. Here we describe a novel mechanism of oncogene activation whereby focal copy number loss of an intronic element within the FTO gene leads to aberrant expression of IRX3, an oncogene in T-cell acute lymphoblastic leukemia (T-ALL). Loss of this CTCF-bound element downstream to IRX3 (+224 kb) leads to enhancer hijack of an upstream developmentally active super-enhancer of the CRNDE long noncoding RNA (-644 kb). Unexpectedly, the CRNDE super-enhancer interacts with the IRX3 promoter with no transcriptional output until it is untethered from the FTO intronic site. We propose that "promoter tethering" of oncogenes to inert regions of the genome is a previously unappreciated biological mechanism preventing tumorigenesis.
Collapse
Affiliation(s)
- Sunniyat Rahman
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Gianna Bloye
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Nadine Farah
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | | | - Joana R Costa
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - David O'Connor
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Rachael Pocock
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Tanya Rapoz-D'Silva
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Adam Turna
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Lingyi Wang
- Department of Developmental Biology and Cancer, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - SooWah Lee
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Adele K Fielding
- Department of Experimental Medicine and Biomedicine, Hull York Medical School, University of York, York, United Kingdom
| | - Juliette Roels
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Roman Jaksik
- Department of Systems Biology and Engineering and Biotechnology Centre, Silesian University of Technology, Gliwice, Poland
| | - Małgorzata Dawidowska
- Department of Molecular and Clinical Genetics, Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | | | - Suzana Hadjur
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
| | - Jim R Hughes
- Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine Centre for Computational Biology, University of Oxford, Oxford, United Kingdom
| | - James O J Davies
- Department of Medicine, Medical Research Council Weatherall Institute of Molecular Medicine Centre for Computational Biology, University of Oxford, Oxford, United Kingdom
| | - Alejandro Gutierrez
- Department of Pediatric Oncology, Dana-Farber/Harvard Cancer Center, Boston, MA
| | - Michelle A Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA
| | - Peter Van Loo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marc R Mansour
- Department of Haematology, University College London Cancer Institute, London, United Kingdom
- Department of Developmental Biology and Cancer, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
5
|
Li Z, Zheng Q, Yang K, Xu T, Wang L, Wang X, Wen W, Wang J, Zhao Y, Song Y, Chen C, Zhou Q, Wu T. CD7 CART Therapy Bridging Allo-HSCT Remarkably Improves Long-Term DFS in Refractory/Relapsed T-ALL/LBL. Transplant Cell Ther 2024:S2666-6367(24)00780-2. [PMID: 39603417 DOI: 10.1016/j.jtct.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/25/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
T-ALL is caused by abnormal proliferation of T cells. It comprises 25%-50% of ALL cases in children and adults. Outlook for R/R T-ALL/LBL and patients over 60 is even dimmer. The treatment is challenging due to its biological and genetic diversity, limiting the development of effective targeted and immunotherapeutic strategies. Salvaged allo-HSCT offers only 20% to 30% DFS. This current study retrospectively analyzed 90 patients with R/R T-ALL (40, 44.4%) or T-LBL (50, 55.6%) treated at Beijing Gobroad Boren Hospital from February 2018 to January 2023. The median age was 14 (range: 2-65) y old. Somatic and germline gene mutations were detected by sequencing pretransplant. Thirty-two (35.6%) patients were sensitive to chemotherapy and achieved CR before transplant (CR group), and 58 (64.4%) cases were resistant to chemotherapy and in non-remission (NR) pre-HSCT. Forty-one of 58 patients in NR received CD7 CAR-T before allo-HSCT (CART group) and the rest 17 patients in NR underwent salvaged transplant (NR group). The results indicate that CD7 CAR-T group have OS (p = .029; 2-y OS rates: 54.4% [95% CI: 38.9% to 76%]) and DFS (p = .00032; 2-y DFS: 51.0% (95% CI: 36.9% to 70.7%)) similar to those in the CR group, but better than those in the NR group. The CIR for CD7 CAR-T group and CR group was significantly lower than NR group after 1 y (p = .0016; CAR-T group 2-y CIR: 31.67% (95% CI: 19.3% to 49.2%)). Our study examined the somatic and germline gene mutations in R/R T-ALL/LBL and evaluated the prognosis after transplantation. Based on our limited study, we found that using CD7 CAR T cells followed by allo-HSCT greatly enhanced the long-term DFS of chemo resistant T-ALL/LBL patients.
Collapse
Affiliation(s)
- Zhihui Li
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Qinlong Zheng
- Department of Medical Laboratory, Beijing Gobroad Boren Hospital, Beijing, China
| | - Keyan Yang
- Department of Medical Laboratory, Beijing Gobroad Boren Hospital, Beijing, China
| | - Teng Xu
- Department of Data Management, Beijing Gobroad Boren Hospital, Beijing, China
| | - Lei Wang
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Xianxuan Wang
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Wiaopei Wen
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Jingjing Wang
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Yongqiang Zhao
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Yanzhi Song
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China
| | - Chen Chen
- Department of Data Management, Beijing Gobroad Boren Hospital, Beijing, China
| | - Qi Zhou
- Department of Data Management, Beijing Gobroad Boren Hospital, Beijing, China
| | - Tong Wu
- Department of Bone Marrow Transplantation, Beijing Gobroad Boren Hospital, Beijing, China.
| |
Collapse
|
6
|
Calvo J, Naguibneva I, Kypraios A, Gilain F, Uzan B, Gaillard B, Bellenger L, Renou L, Antoniewski C, Lapillonne H, Petit A, Ballerini P, Mancini SJ, Marchand T, Peyron JF, Pflumio F. High CD44 expression and enhanced E-selectin binding identified as biomarkers of chemoresistant leukemic cells in human T-ALL. Leukemia 2024:10.1038/s41375-024-02473-7. [PMID: 39580584 DOI: 10.1038/s41375-024-02473-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a hematopoietic malignancy characterized by increased proliferation and incomplete maturation of T-cell progenitors, for which relapse is often of poor prognosis. To improve patient outcomes, it is critical to understand the chemoresistance mechanisms arising from cell plasticity induced by the bone marrow (BM) microenvironment. Single-cell RNA sequencing of human T-ALL cells from adipocyte-rich and adipocyte-poor BM revealed a distinct leukemic cell population defined by quiescence and high CD44 expression (Ki67neg/lowCD44high). During in vivo treatment, these cells evaded chemotherapy, and were further called Chemotherapy-resistant Leukemic Cells (CLCs). Patient sample analysis revealed Ki67neg/lowCD44high CLCs at diagnosis and during relapse, with each displaying a specific transcriptomic signature. Interestingly, CD44high expression in T-ALL Ki67neg/low CLCs was associated with E-selectin binding. Analysis of 39 human T-ALL samples revealed significantly enhanced E-selectin binding activity in relapse/refractory samples compared with drug-sensitive samples. These characteristics of chemoresistant T-ALL CLCs provide key insights for prognostic stratification and novel therapeutic options.
Collapse
Affiliation(s)
- Julien Calvo
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France.
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France.
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France.
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France.
| | - Irina Naguibneva
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France
| | - Anthony Kypraios
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, 06204, Nice, France
| | - Florian Gilain
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| | - Benjamin Uzan
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France
| | - Baptiste Gaillard
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France
| | - Lea Bellenger
- ARTbio Bioinformatics Analysis Facility, IBPS, CNRS, Sorbonne Université, Institut Français de Bioinformatique, 75005, Paris, France
| | - Laurent Renou
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| | - Christophe Antoniewski
- ARTbio Bioinformatics Analysis Facility, IBPS, CNRS, Sorbonne Université, Institut Français de Bioinformatique, 75005, Paris, France
| | - Helene Lapillonne
- Sorbonne University, AP-HP, Laboratory of Hematology, Armand-Trousseau Hospital, 75012, Paris, France
- Sorbonne Université, Centre de Recherche Saint-Antoine UMR_S938, Pediatric Hematology Oncology Unit, AP-HP, Armand-Trousseau Hospital, 75012, Paris, France
| | - Arnaud Petit
- Sorbonne University, AP-HP, Laboratory of Hematology, Armand-Trousseau Hospital, 75012, Paris, France
- Sorbonne Université, Centre de Recherche Saint-Antoine UMR_S938, Pediatric Hematology Oncology Unit, AP-HP, Armand-Trousseau Hospital, 75012, Paris, France
| | - Paola Ballerini
- Sorbonne University, AP-HP, Laboratory of Hematology, Armand-Trousseau Hospital, 75012, Paris, France
- Sorbonne Université, Centre de Recherche Saint-Antoine UMR_S938, Pediatric Hematology Oncology Unit, AP-HP, Armand-Trousseau Hospital, 75012, Paris, France
| | | | - Tony Marchand
- Université Rennes, EFS, Inserm, MOBIDIC-UMR_S 1236, F-35000, Rennes, France
- Service d'hématologie Clinique, Centre Hospitalier Universitaire de Rennes, 35003, Rennes, France
| | - Jean-François Peyron
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), INSERM U1065, 06204, Nice, France
| | - Françoise Pflumio
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/SGCSR/Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), F-92260, Fontenay-aux-Roses, France
- Laboratoire des cellules Souches Hématopoïétiques et des Leucémies, Equipe Niche et Cancer dans l'Hématopoïèse, équipe labellisée Ligue Nationale Contre le Cancer, Unité Mixte de Recherche (UMR) 1274-E008, Inserm, CEA, 92265, Fontenay-aux Roses, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| |
Collapse
|
7
|
Roy U, Sharma A, Sharma S, Dahal S, Kumari N, Desai SS, Kumari S, Dixit J, Sharma M A, Nujoom N, Choudhary B, Raghavan SC. Mutations at BCL11B Exon 4 Associated with T Cell Acute Lymphoblastic Leukemia Are Facilitated by AID and Formation of Non-B DNA Conformations. Mol Cell Biol 2024; 44:590-606. [PMID: 39511874 PMCID: PMC11583620 DOI: 10.1080/10985549.2024.2419661] [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: 04/11/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
One of the primary reasons behind the pathogenesis of T cell acute lymphoblastic leukemia (T-ALL) is the deregulation of the transcription factor BCL11B. The exon 4 of BCL11B harbors several driver mutations, which abolishes its DNA-binding ability. The high frequency of C > T or G > A conversion in close vicinity of AID (Activation-induced cytidine deaminase)-hotspot motifs in the deregulated gene prompted us to investigate the role of AID in BCL11B mutagenesis. Our results reveal that AID is expressed in T-ALL patient-derived cells, binds to BCL11B fragile region (FR) in exon 4 of T cells in vivo, and generates a signature mutation pattern in this region. The mutation frequency in BCL11B FR could be modulated upon overexpression of the AID gene in the knockout background, further suggesting the involvement of AID in BCL11B mutagenesis. Importantly, various lines of experimentation reveal that BCL11B FR could fold into parallel G-quadruplex, triplex, and hairpin structures, which could act as a replication/transcription block, causing mutagenesis. Thus, our results suggest that AID binds to BCL11B exon 4 due to non-B DNA formation, causing U:G mismatches or replication blocks, which, when repaired erroneously, generates deleterious mutations, resulting in loss of functionality of BCL11B, and thus becomes the cause of T-ALL.
Collapse
Affiliation(s)
- Urbi Roy
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Anju Sharma
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Shivangi Sharma
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Sumedha Dahal
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Nitu Kumari
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | - Susmita Kumari
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Jyotika Dixit
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Arun Sharma M
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Najma Nujoom
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| | | |
Collapse
|
8
|
Lyu A, Nam SH, Humphrey RS, Horton TM, Ehrlich LIR. Cells and signals of the leukemic microenvironment that support progression of T-cell acute lymphoblastic leukemia (T-ALL). Exp Mol Med 2024; 56:2337-2347. [PMID: 39482533 PMCID: PMC11612169 DOI: 10.1038/s12276-024-01335-7] [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: 06/06/2024] [Revised: 07/30/2024] [Accepted: 08/11/2024] [Indexed: 11/03/2024] Open
Abstract
Current intensified chemotherapy regimens have significantly increased survival rates for pediatric patients with T-cell acute lymphoblastic leukemia (T-ALL), but these treatments can result in serious adverse effects; furthermore, patients who are resistant to chemotherapy or who relapse have inferior outcomes, together highlighting the need for improved therapeutic strategies. Despite recent advances in stratifying T-ALL into molecular subtypes with distinct driver mutations, efforts to target the tumor-intrinsic genomic alterations critical for T-ALL progression have yet to translate into more effective and less toxic therapies. Ample evidence now indicates that extrinsic factors in the leukemic microenvironment are critical for T-ALL growth, infiltration, and therapeutic resistance. Considering the diversity of organs infiltrated by T-ALL cells and the unique cellular components of the microenvironment encountered at each site, it is likely that there are both shared features of tumor-supportive niches across multiple organs and site-specific features that are key to leukemia cell survival. Therefore, elucidating the distinct microenvironmental cues supporting T-ALL in different anatomic locations could reveal novel therapeutic targets to improve therapies. This review summarizes the current understanding of the intricate interplay between leukemia cells and the diverse cells they encounter within their tumor microenvironments (TMEs), as well as opportunities to therapeutically target the leukemic microenvironment.
Collapse
Affiliation(s)
- Aram Lyu
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Seo Hee Nam
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Ryan S Humphrey
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Terzah M Horton
- Department of Pediatrics, Baylor College of Medicine/Dan L. Duncan Cancer Center and Texas Children's Cancer Center, Houston, TX, USA
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA.
- Department of Oncology, Livestrong Cancer Institutes, The University of Texas at Austin Dell Medical School, Austin, TX, USA.
| |
Collapse
|
9
|
Aertgeerts M, Meyers S, Demeyer S, Segers H, Cools J. Unlocking the Complexity: Exploration of Acute Lymphoblastic Leukemia at the Single Cell Level. Mol Diagn Ther 2024; 28:727-744. [PMID: 39190087 DOI: 10.1007/s40291-024-00739-5] [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] [Accepted: 08/08/2024] [Indexed: 08/28/2024]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common cancer in children. ALL originates from precursor lymphocytes that acquire multiple genomic changes over time, including chromosomal rearrangements and point mutations. While a large variety of genomic defects was identified and characterized in ALL over the past 30 years, it was only in recent years that the clonal heterogeneity was recognized. Thanks to the latest advancements in single-cell sequencing techniques, which have evolved from the analysis of a few hundred cells to the analysis of thousands of cells simultaneously, the study of tumor heterogeneity now becomes possible. Different modalities can be explored at the single-cell level: DNA, RNA, epigenetic modifications, and intracellular and cell surface proteins. In this review, we describe these techniques and highlight their advantages and limitations in the study of ALL biology. Moreover, multiomics technologies and the incorporation of the spatial dimension can provide insight into intercellular communication. We describe how the different single-cell sequencing technologies help to unravel the molecular complexity of ALL, shedding light on its development, its heterogeneity, its interaction with the leukemia microenvironment and possible relapse mechanisms.
Collapse
Affiliation(s)
- Margo Aertgeerts
- Department of Oncology, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven-UZ Leuven, Leuven, Belgium
| | - Sarah Meyers
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven-UZ Leuven, Leuven, Belgium
| | - Sofie Demeyer
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Leuvens Kanker Instituut (LKI), KU Leuven-UZ Leuven, Leuven, Belgium
| | - Heidi Segers
- Department of Oncology, KU Leuven, Leuven, Belgium.
- Leuvens Kanker Instituut (LKI), KU Leuven-UZ Leuven, Leuven, Belgium.
- Department of Pediatric Hematology and Oncology, UZ Leuven, Leuven, Belgium.
| | - Jan Cools
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Center for Cancer Biology, VIB, Leuven, Belgium.
- Leuvens Kanker Instituut (LKI), KU Leuven-UZ Leuven, Leuven, Belgium.
| |
Collapse
|
10
|
Meyers S, Gielen O, Cools J, Demeyer S. Single-cell CRISPR screening characterizes transcriptional deregulation in T-cell acute lymphoblastic leukemia. Haematologica 2024; 109:3167-3181. [PMID: 38813729 PMCID: PMC11443379 DOI: 10.3324/haematol.2023.284901] [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: 12/27/2023] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive type of leukemia caused by accumulation of multiple genetic alterations in T-cell progenitors. However, for many genes it remains unknown how their mutations contribute to disease development. We therefore performed two single-cell CRISPR screens in primary pro-T cells ex vivo to study the transcriptional impact of loss-of-function alterations in T-ALL and correlate this with effects on cell fitness. The various perturbations were clustered based on their effects on E2F/MYC or STAT/NOTCH signatures, which play a defining role in driving T-cell proliferation. Many of the perturbations resulted in positive effects on the STAT and NOTCH signatures and were predicted to behave as haploinsufficient tumor suppressors in T-ALL. Additionally, Spi1 was identified as an essential gene for pro-T-cell survival, associated with deregulation of the MYC signature and epigenetic consequences. In contrast, Bcl11b was identified as a strong tumor suppressor gene in immature T lymphocytes, associated with deregulation of NF-kB and JAK/STAT signaling. We found a correlation between BCL11B expression level and JAK/STAT pathway mutations in T-ALL patients and demonstrated oncogenic cooperation between Bcl11b inactivation and JAK3 hyperactivation in pro-T cells. Altogether, these single-cell CRISPR screens in pro-T cells provide fundamental insights into the mechanisms of transcriptional deregulation caused by genetic alterations in T-ALL.
Collapse
Affiliation(s)
- Sarah Meyers
- Center for Human Genetics, KU Leuven, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium; Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven
| | - Olga Gielen
- Center for Human Genetics, KU Leuven, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium; Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven
| | - Jan Cools
- Center for Human Genetics, KU Leuven, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium; Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven.
| | - Sofie Demeyer
- Center for Human Genetics, KU Leuven, Leuven, Belgium; Center for Cancer Biology, VIB, Leuven, Belgium; Leuvens Kanker Instituut (LKI), KU Leuven - UZ Leuven, Leuven.
| |
Collapse
|
11
|
Risinskaya N, Abdulpatakhov A, Chabaeva Y, Aleshina O, Gladysheva M, Nikulina E, Bolshakov I, Yushkova A, Dubova O, Vasileva A, Obukhova T, Julhakyan H, Kapranov N, Galtseva I, Kulikov S, Sudarikov A, Parovichnikova E. Biallelic Loss of 7q34 ( TRB) and 9p21.3 ( CDKN2A/ 2B) in Adult Ph-Negative Acute T-Lymphoblastic Leukemia. Int J Mol Sci 2024; 25:10482. [PMID: 39408811 PMCID: PMC11477120 DOI: 10.3390/ijms251910482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/16/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Tumor cells of acute lymphoblastic leukemia (ALL) may have various genetic abnormalities. Some of them lead to a complete loss of certain genes. Our aim was to reveal biallelic deletions of genes in Ph-negative T-ALL. Chromosomal microarray analysis (CMA) was performed for 47 patients with de novo Ph-negative T-ALL, who received treatment according to RALL-2016m clinical protocol at the National Medical Research Center for Hematology (Moscow, Russia) from 2017 to 2023. Out of forty-seven patients, only three had normal molecular karyotype. The other 44 patients had multiple gains, losses, and copy neutral losses of heterozygosity. Biallelic losses were found in 14 patients (30%). In ten patients (21%), a biallelic deletion of 9p21.3 involved a different number of genes, however CDKN2A gene loss was noted in all ten cases. For seven patients (15%), a biallelic deletion of 7q34 was found, including two genes-PRSS1, PRSS2 located within the T-cell receptor beta (TRB) locus. A clonal rearrangement of the TRB gene was revealed in 6 out of 7 cases with 7q34 biallelic loss. Both biallelic deletions can be considered favorable prognostic factors, with an association with 9p21 being statistically significant (p = 0.01) and a trend for 7q34 (p = 0.12) being observed.
Collapse
Affiliation(s)
- Natalya Risinskaya
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Abdulpatakh Abdulpatakhov
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Yulia Chabaeva
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Olga Aleshina
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Maria Gladysheva
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Elena Nikulina
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Ivan Bolshakov
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Anna Yushkova
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Olga Dubova
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
- Institute of Biodesign and Modeling of Complex Systems, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Anastasia Vasileva
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Tatiana Obukhova
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Hunan Julhakyan
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Nikolay Kapranov
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Irina Galtseva
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Sergey Kulikov
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Andrey Sudarikov
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| | - Elena Parovichnikova
- National Medical Research Center for Hematology, 125167 Moscow, Russia; (A.A.); (Y.C.); (O.A.); (M.G.); (E.N.); (I.B.); (A.Y.); (O.D.); (A.V.); (T.O.); (H.J.); (N.K.); (I.G.); (S.K.); (A.S.)
| |
Collapse
|
12
|
Besser E, Gelfand A, Procaccia S, Berman P, Meiri D. Cannabinoid combination targets NOTCH1-mutated T-cell acute lymphoblastic leukemia through the integrated stress response pathway. eLife 2024; 12:RP90854. [PMID: 39258755 PMCID: PMC11390110 DOI: 10.7554/elife.90854] [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] [Indexed: 09/12/2024] Open
Abstract
In T-cell acute lymphoblastic leukemia (T-ALL), more than 50% of cases display autoactivation of Notch1 signaling, leading to oncogenic transformation. We have previously identified a specific chemovar of Cannabis that induces apoptosis by preventing Notch1 maturation in leukemia cells. Here, we isolated three cannabinoids from this chemovar that synergistically mimic the effects of the whole extract. Two were previously known, cannabidiol (CBD) and cannabidivarin (CBDV), whereas the third cannabinoid, which we termed 331-18A, was identified and fully characterized in this study. We demonstrated that these cannabinoids act through cannabinoid receptor type 2 and TRPV1 to activate the integrated stress response pathway by depleting intracellular Ca2+. This is followed by increased mRNA and protein expression of ATF4, CHOP, and CHAC1, which is hindered by inhibiting the upstream initiation factor eIF2α. The increased abundance of CHAC1 prevents Notch1 maturation, thereby reducing the levels of the active Notch1 intracellular domain, and consequently decreasing cell viability and increasing apoptosis. Treatment with the three isolated molecules resulted in reduced tumor size and weight in vivo and slowed leukemia progression in mice models. Altogether, this study elucidated the mechanism of action of three distinct cannabinoids in modulating the Notch1 pathway, and constitutes an important step in the establishment of a new therapy for treating NOTCH1-mutated diseases and cancers such as T-ALL.
Collapse
Affiliation(s)
- Elazar Besser
- The Laboratory of Cancer Biology and Cannabinoid Research, Faculty of Biology, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Anat Gelfand
- The Laboratory of Cancer Biology and Cannabinoid Research, Faculty of Biology, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Shiri Procaccia
- The Laboratory of Cancer Biology and Cannabinoid Research, Faculty of Biology, Technion – Israel Institute of TechnologyHaifaIsrael
| | - Paula Berman
- The Laboratory of Cancer Biology and Cannabinoid Research, Faculty of Biology, Technion – Israel Institute of TechnologyHaifaIsrael
| | - David Meiri
- The Laboratory of Cancer Biology and Cannabinoid Research, Faculty of Biology, Technion – Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
13
|
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
|
14
|
Le Maout C, Fahy L, Renou L, Devanand C, Duwat C, Barroca V, Le Gall M, Ballerini P, Petit A, Calvo J, Uzan B, Pflumio F, Petit V. T-cell acute lymphoblastic leukemia progression is supported by inflammatory molecules including hepatocyte growth factor. Biomed Pharmacother 2024; 177:117039. [PMID: 38955085 DOI: 10.1016/j.biopha.2024.117039] [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: 04/30/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematological disorder characterized by an increased proliferation of immature T lymphocytes precursors. T-ALL treatment includes chemotherapy with strong side effects, and patients that undergo relapse display poor prognosis. Although cell-intrinsic oncogenic pathways are well-studied, the tumor microenvironment, like inflammatory cellular and molecular components is less explored in T-ALL. We sought to determine the composition of the inflammatory microenvironment induced by T-ALL, and its role in T-ALL progression. We show in two mouse T-ALL cell models that T-ALLs enhance blood neutrophils and resident monocytes, accompanied with a plasmatic acute secretion of inflammatory molecules. Depleting neutrophils using anti-Ly6G treatment or resident monocytes by clodronate liposomes treatment does not modulate plasmatic inflammatory molecule secretion and mice survival. However, inhibiting the secretion of inflammatory molecules by microenvironment with NECA, an agonist of adenosine receptors, diminishes T-ALL progression enhancing mouse survival. We uncovered Hepatocyte Growth Factor (HGF), T-ALL-driven and the most decreased molecule with NECA, as a potential therapeutic target in T-ALL. Altogether, we identified a signature of inflammatory molecules that can potentially be involved in T-ALL evolution and uncovered HGF/cMET pathway as important to target for limiting T-ALL progression.
Collapse
Affiliation(s)
- Charly Le Maout
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France
| | - Lucine Fahy
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France
| | - Laurent Renou
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France
| | - Caroline Devanand
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France
| | - Charlotte Duwat
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France
| | - Vilma Barroca
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France
| | - Morgane Le Gall
- Proteom'IC facility, Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris F-75014, France
| | - Paola Ballerini
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris, Hôpital A. Trousseau, Paris, France
| | - Arnaud Petit
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris, Hôpital A. Trousseau, Paris, France
| | - Julien Calvo
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France; Institut Carnot OPALE, Hôpital Saint Louis, Paris F-75020, France
| | - Benjamin Uzan
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France; Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris F-75013, France
| | - Françoise Pflumio
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France; Institut Carnot OPALE, Hôpital Saint Louis, Paris F-75020, France.
| | - Vanessa Petit
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France; Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), France.
| |
Collapse
|
15
|
Lahera A, Vela-Martín L, Fernández-Navarro P, Llamas P, López-Lorenzo JL, Cornago J, Santos J, Fernández-Piqueras J, Villa-Morales M. PIM1 is a potential therapeutic target for the leukemogenic effects mediated by JAK/STAT pathway mutations in T-ALL/LBL. NPJ Precis Oncol 2024; 8:152. [PMID: 39033228 PMCID: PMC11271448 DOI: 10.1038/s41698-024-00638-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
Precursor T-cell neoplasms (T-ALL/LBL) are aggressive hematological malignancies that arise from the malignant transformation of immature thymocytes. Despite the JAK/STAT pathway is recurrently altered in these neoplasms, there are not pharmacological inhibitors officially approved for the treatment of T-ALL/LBL patients that present oncogenic JAK/STAT pathway mutations. In the effort to identify potential therapeutic targets for those patients, we followed an alternative approach and focused on their transcriptional profile. We combined the analysis of molecular data from T-ALL/LBL patients with the generation of hematopoietic cellular models to reveal that JAK/STAT pathway mutations are associated with an aberrant transcriptional profile. Specifically, we demonstrate that JAK/STAT pathway mutations induce the overexpression of the PIM1 gene. Moreover, we show that the pan-PIM inhibitor, PIM447, significantly reduces the leukemogenesis, as well as the aberrant activation of c-MYC and mTOR pathways in cells expressing different JAK/STAT pathway mutations, becoming a potential therapeutic opportunity for a relevant subset of T-ALL/LBL patients.
Collapse
Affiliation(s)
- Antonio Lahera
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain.
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain.
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain.
| | - Laura Vela-Martín
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Pablo Fernández-Navarro
- Unit of Cancer and Environmental Epidemiology, Centro Nacional de Epidemiología, Instituto de Salud Carlos III, Madrid, 28029, Spain
- Consorcio de Investigación Biomédica de Epidemiología y Salud Pública (CIBERESP), Madrid, 28029, Spain
| | - Pilar Llamas
- Division of Hematology and Hemotherapy, Hospital Universitario Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - José L López-Lorenzo
- Division of Hematology and Hemotherapy, Hospital Universitario Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Javier Cornago
- Division of Hematology and Hemotherapy, Hospital Universitario Fundación Jiménez Díaz, Madrid, 28040, Spain
| | - Javier Santos
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid) Madrid, Madrid, 28049, Spain
| | - José Fernández-Piqueras
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain.
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain.
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain.
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid) Madrid, Madrid, 28049, Spain.
| | - María Villa-Morales
- Department of Biology, Universidad Autónoma de Madrid, Madrid, 28049, Spain.
- Department of Genome dynamics and function, Centro de Biología Molecular Severo Ochoa (CBM), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, 28049, Spain.
- Area of Genetics and Genomics, IIS Fundación Jiménez Díaz, Madrid, 28040, Spain.
- Institute for Molecular Biology-IUBM (Universidad Autónoma de Madrid) Madrid, Madrid, 28049, Spain.
| |
Collapse
|
16
|
Lundgren S, Myllymäki M, Järvinen T, Keränen MAI, Theodoropoulos J, Smolander J, Kim D, Salmenniemi U, Walldin G, Savola P, Kelkka T, Rajala H, Hellström-Lindberg E, Itälä-Remes M, Kankainen M, Mustjoki S. Somatic mutations associate with clonal expansion of CD8 + T cells. SCIENCE ADVANCES 2024; 10:eadj0787. [PMID: 38848368 PMCID: PMC11160466 DOI: 10.1126/sciadv.adj0787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 05/06/2024] [Indexed: 06/09/2024]
Abstract
Somatic mutations in T cells can cause cancer but also have implications for immunological diseases and cell therapies. The mutation spectrum in nonmalignant T cells is unclear. Here, we examined somatic mutations in CD4+ and CD8+ T cells from 90 patients with hematological and immunological disorders and used T cell receptor (TCR) and single-cell sequencing to link mutations with T cell expansions and phenotypes. CD8+ cells had a higher mutation burden than CD4+ cells. Notably, the biggest variant allele frequency (VAF) of non-synonymous variants was higher than synonymous variants in CD8+ T cells, indicating non-random occurrence. The non-synonymous VAF in CD8+ T cells strongly correlated with the TCR frequency, but not age. We identified mutations in pathways essential for T cell function and often affected lymphoid neoplasia. Single-cell sequencing revealed cytotoxic TEMRA phenotypes of mutated T cells. Our findings suggest that somatic mutations contribute to CD8+ T cell expansions without malignant transformation.
Collapse
Affiliation(s)
- Sofie Lundgren
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Timo Järvinen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko A. I. Keränen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Jason Theodoropoulos
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Johannes Smolander
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Daehong Kim
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Urpu Salmenniemi
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Stem Cell Transplantation Unit, Turku University Hospital, Turku, Finland
| | - Gunilla Walldin
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Paula Savola
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Clinical Chemistry, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tiina Kelkka
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Hanna Rajala
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Maija Itälä-Remes
- Stem Cell Transplantation Unit, Turku University Hospital, Turku, Finland
| | - Matti Kankainen
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
- ICAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
17
|
Xu J, Peng J, Sun S, Wang D, Yuan W, Yang X, Shi T, Wang R, Liu H, Zhang P, Zhu HH. Preclinical testing of CT1113, a novel USP28 inhibitor, for the treatment of T-cell acute lymphoblastic leukaemia. Br J Haematol 2024; 204:2301-2318. [PMID: 38685813 DOI: 10.1111/bjh.19492] [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: 11/04/2023] [Accepted: 04/16/2024] [Indexed: 05/02/2024]
Abstract
T-cell acute lymphoblastic leukaemia (T-ALL) is a highly aggressive and heterogeneous lymphoid malignancy with poor prognosis in adult patients. Aberrant activation of the NOTCH1 signalling pathway is involved in the pathogenesis of over 60% of T-ALL cases. Ubiquitin-specific protease 28 (USP28) is a deubiquitinase known to regulate the stability of NOTCH1. Here, we report that genetic depletion of USP28 or using CT1113, a potent small molecule targeting USP28, can strongly destabilize NOTCH1 and inhibit the growth of T-ALL cells. Moreover, we show that USP28 also regulates the stability of sterol regulatory element binding protein 1 (SREBP1), which has been reported to mediate increased lipogenesis in tumour cells. As the most critical transcription factor involved in regulating lipogenesis, SREBP1 plays an important role in the metabolism of T-ALL. Therefore, USP28 may be a potential therapeutic target, and CT1113 may be a promising novel drug for T-ALL with or without mutant NOTCH1.
Collapse
Affiliation(s)
- Jieyu Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Jin Peng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shu Sun
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, China
| | - Donghai Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Wei Yuan
- Department of Hematology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Xueying Yang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ting Shi
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Rong Wang
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Hudan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, China
| | - Pumin Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Translational Medicine, Zhejiang University Medical School, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong-Hu Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| |
Collapse
|
18
|
Cardoso BA, Duque M, Gírio A, Fragoso R, Oliveira ML, Allen JR, Martins LR, Correia NC, Silveira AB, Veloso A, Kimura S, Demoen L, Matthijssens F, Jeha S, Cheng C, Pui CH, Grosso AR, Neto JL, De Almeida SF, Van Vlieberghe P, Mullighan CG, Yunes JA, Langenau DM, Pflumio F, Barata JT. CASZ1 upregulates PI3K-AKT-mTOR signaling and promotes T-cell acute lymphoblastic leukemia. Haematologica 2024; 109:1713-1725. [PMID: 38058200 PMCID: PMC11141679 DOI: 10.3324/haematol.2023.282854] [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: 02/10/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
CASZ1 is a conserved transcription factor involved in neural development, blood vessel assembly and heart morphogenesis. CASZ1 has been implicated in cancer, either suppressing or promoting tumor development depending on the tissue. However, the impact of CASZ1 on hematological tumors remains unknown. Here, we show that the T-cell oncogenic transcription factor TAL1 is a direct positive regulator of CASZ1, that T-cell acute lymphoblastic leukemia (T-ALL) samples at diagnosis overexpress CASZ1b isoform, and that CASZ1b expression in patient samples correlates with PI3K-AKT-mTOR signaling pathway activation. In agreement, overexpression of CASZ1b in both Ba/F3 and T-ALL cells leads to the activation of PI3K signaling pathway, which is required for CASZ1b-mediated transformation of Ba/F3 cells in vitro and malignant expansion in vivo. We further demonstrate that CASZ1b cooperates with activated NOTCH1 to promote T-ALL development in zebrafish, and that CASZ1b protects human T-ALL cells from serum deprivation and treatment with chemotherapeutic drugs. Taken together, our studies indicate that CASZ1b is a TAL1-regulated gene that promotes T-ALL development and resistance to chemotherapy.
Collapse
Affiliation(s)
- Bruno A Cardoso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Mafalda Duque
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Ana Gírio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Rita Fragoso
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Mariana L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - James R Allen
- MGH Pathology and Harvard Medical School, Charlestown MA 02129
| | - Leila R Martins
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Nádia C Correia
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | | | | | - Shunsuke Kimura
- Department of Pathology, Center of Excellence for Leukemia Studies, and Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis TN
| | - Lisa Demoen
- Department of Biomolecular Medicine, Ghent University, and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Filip Matthijssens
- Department of Biomolecular Medicine, Ghent University, and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Sima Jeha
- Department of Oncology, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis TN, US; Department of Global Pediatric Medicine, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis TN
| | - Cheng Cheng
- Department of Biostatistics, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis TN
| | - Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis TN, US; Department of Global Pediatric Medicine, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis TN, US; Department of Pathology, St. Jude Children's Research Hospital and the University of Tennessee Health Science Center, Memphis TN
| | - Ana R Grosso
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica
| | - João L Neto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Sérgio F De Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Pieter Van Vlieberghe
- Department of Biomolecular Medicine, Ghent University, and Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Charles G Mullighan
- Department of Pathology, Center of Excellence for Leukemia Studies, and Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis TN
| | - J Andres Yunes
- Laboratório de Biologia Molecular, Centro Infantil Boldrini, Campinas, SP
| | | | - Françoise Pflumio
- Université Paris-Saclay, INSERM, iRCM/IBFJ CEA, UMR Stabilité Génétique Cellules Souches et Radiations, F-92265, Fontenay-aux-Roses, France; OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Saint-Louis Hospital, 75010 Paris
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon.
| |
Collapse
|
19
|
Lizcova L, Prihodova E, Pavlistova L, Svobodova K, Mejstrikova E, Hrusak O, Luknarova P, Janotova I, Sramkova L, Stary J, Zemanova Z. Cytogenomic characterization of pediatric T-cell acute lymphoblastic leukemia reveals TCR rearrangements as predictive factors for exceptional prognosis. Mol Cytogenet 2024; 17:14. [PMID: 38783324 PMCID: PMC11118568 DOI: 10.1186/s13039-024-00682-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND T-cell acute lymphoblastic leukemia (T-ALL) represents a rare and clinically and genetically heterogeneous disease that constitutes 10-15% of newly diagnosed pediatric ALL cases. Despite improved outcomes of these children, the survival rate after relapse is extremely poor. Moreover, the survivors must also endure the acute and long-term effects of intensive therapy. Although recent studies have identified a number of recurrent genomic aberrations in pediatric T-ALL, none of the changes is known to have prognostic significance. The aim of our study was to analyze the cytogenomic changes and their various combinations in bone marrow cells of children with T-ALL and to correlate our findings with the clinical features of the subjects and their treatment responses. RESULTS We performed a retrospective and prospective comprehensive cytogenomic analysis of consecutive cohort of 66 children (46 boys and 20 girls) with T-ALL treated according to BFM-based protocols and centrally investigated cytogenetics and immunophenotypes. Using combinations of cytogenomic methods (conventional cytogenetics, FISH, mFISH/mBAND, arrayCGH/SNP and MLPA), we identified chromosomal aberrations in vast majority of patients (91%). The most frequent findings involved the deletion of CDKN2A/CDKN2B genes (71%), T-cell receptor (TCR) loci translocations (27%), and TLX3 gene rearrangements (23%). All chromosomal changes occurred in various combinations and were rarely found as a single abnormality. Children with aberrations of TCR loci had a significantly better event free (p = 0.0034) and overall survival (p = 0.0074), all these patients are living in the first complete remission. None of the abnormalities was an independent predictor of an increased risk of relapse. CONCLUSIONS We identified a subgroup of patients with TCR aberrations (both TRA/TRD and TRB), who had an excellent prognosis in our cohort with 5-year EFS and OS of 100%, regardless of the presence of other abnormality or the translocation partner. Our data suggest that escalation of treatment intensity, which may be considered in subsets of T-ALL is not needed for nonHR (non-high risk) patients with TCR aberrations.
Collapse
Affiliation(s)
- Libuse Lizcova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic.
| | - Eva Prihodova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Lenka Pavlistova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Karla Svobodova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Ester Mejstrikova
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Ondrej Hrusak
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Pavla Luknarova
- CLIP - Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Iveta Janotova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Lucie Sramkova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague and University Hospital Motol, Prague, Czech Republic
| | - Zuzana Zemanova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital in Prague and First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
20
|
Cao X, Huber S, Ahari AJ, Traube FR, Seifert M, Oakes CC, Secheyko P, Vilov S, Scheller IF, Wagner N, Yépez VA, Blombery P, Haferlach T, Heinig M, Wachutka L, Hutter S, Gagneur J. Analysis of 3760 hematologic malignancies reveals rare transcriptomic aberrations of driver genes. Genome Med 2024; 16:70. [PMID: 38769532 PMCID: PMC11103968 DOI: 10.1186/s13073-024-01331-6] [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: 09/29/2023] [Accepted: 04/04/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Rare oncogenic driver events, particularly affecting the expression or splicing of driver genes, are suspected to substantially contribute to the large heterogeneity of hematologic malignancies. However, their identification remains challenging. METHODS To address this issue, we generated the largest dataset to date of matched whole genome sequencing and total RNA sequencing of hematologic malignancies from 3760 patients spanning 24 disease entities. Taking advantage of our dataset size, we focused on discovering rare regulatory aberrations. Therefore, we called expression and splicing outliers using an extension of the workflow DROP (Detection of RNA Outliers Pipeline) and AbSplice, a variant effect predictor that identifies genetic variants causing aberrant splicing. We next trained a machine learning model integrating these results to prioritize new candidate disease-specific driver genes. RESULTS We found a median of seven expression outlier genes, two splicing outlier genes, and two rare splice-affecting variants per sample. Each category showed significant enrichment for already well-characterized driver genes, with odds ratios exceeding three among genes called in more than five samples. On held-out data, our integrative modeling significantly outperformed modeling based solely on genomic data and revealed promising novel candidate driver genes. Remarkably, we found a truncated form of the low density lipoprotein receptor LRP1B transcript to be aberrantly overexpressed in about half of hairy cell leukemia variant (HCL-V) samples and, to a lesser extent, in closely related B-cell neoplasms. This observation, which was confirmed in an independent cohort, suggests LRP1B as a novel marker for a HCL-V subclass and a yet unreported functional role of LRP1B within these rare entities. CONCLUSIONS Altogether, our census of expression and splicing outliers for 24 hematologic malignancy entities and the companion computational workflow constitute unique resources to deepen our understanding of rare oncogenic events in hematologic cancers.
Collapse
Affiliation(s)
- Xueqi Cao
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Graduate School of Quantitative Biosciences (QBM), Munich, Germany
| | - Sandra Huber
- Munich Leukemia Laboratory (MLL), Munich, Germany
| | - Ata Jadid Ahari
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Franziska R Traube
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Stuttgart, Germany
| | - Marc Seifert
- Department of Haematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christopher C Oakes
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Polina Secheyko
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Faculty of Biology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Sergey Vilov
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Ines F Scheller
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Nils Wagner
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Helmholtz Association - Munich School for Data Science (MUDS), Munich, Germany
| | - Vicente A Yépez
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Piers Blombery
- Peter MacCallum Cancer Centre, Melbourne, Australia
- University of Melbourne, Melbourne, Australia
- Torsten Haferlach Leukämiediagnostik Stiftung, Munich, Germany
| | | | - Matthias Heinig
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Leonhard Wachutka
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
| | | | - Julien Gagneur
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
- Graduate School of Quantitative Biosciences (QBM), Munich, Germany.
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany.
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany.
| |
Collapse
|
21
|
De Coninck S, De Smedt R, Lintermans B, Reunes L, Kosasih HJ, Reekmans A, Brown LM, Van Roy N, Palhais B, Roels J, Van der Linden M, Van Dorpe J, Ntziachristos P, Van Delft FW, Mansour MR, Pieters T, Lammens T, De Moerloose B, De Bock CE, Goossens S, Van Vlierberghe P. Targeting hyperactive platelet-derived growth factor receptor-β signaling in T-cell acute lymphoblastic leukemia and lymphoma. Haematologica 2024; 109:1373-1384. [PMID: 37941480 PMCID: PMC11063843 DOI: 10.3324/haematol.2023.283981] [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: 08/17/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) and T-cell lymphoblastic lymphoma (T-LBL) are rare aggressive hematologic malignancies. Current treatment consists of intensive chemotherapy leading to 80% overall survival but is associated with severe toxic side effects. Furthermore, 10-20% of patients still die from relapsed or refractory disease providing a strong rationale for more specific, targeted therapeutic strategies with less toxicities. Here, we report a novel MYH9::PDGFRB fusion in a T-LBL patient, and demonstrate that this fusion product is constitutively active and sufficient to drive oncogenic transformation in vitro and in vivo. Expanding our analysis more broadly across T-ALL, we found a T-ALL cell line and multiple patient-derived xenograft models with PDGFRB hyperactivation in the absence of a fusion, with high PDGFRB expression in TLX3 and HOXA T-ALL molecular subtypes. To target this PDGFRB hyperactivation, we evaluated the therapeutic effects of a selective PDGFRB inhibitor, CP-673451, both in vitro and in vivo and demonstrated sensitivity if the receptor is hyperactivated. Altogether, our work reveals that hyperactivation of PDGFRB is an oncogenic driver in T-ALL/T-LBL, and that screening T-ALL/T-LBL patients for phosphorylated PDGFRB levels can serve as a biomarker for PDGFRB inhibition as a novel targeted therapeutic strategy in their treatment regimen.
Collapse
Affiliation(s)
- Stien De Coninck
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent
| | - Renate De Smedt
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent
| | - Beatrice Lintermans
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent
| | - Lindy Reunes
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent
| | - Hansen J Kosasih
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia; School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW
| | - Alexandra Reekmans
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent
| | - Lauren M Brown
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia; School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW
| | - Nadine Van Roy
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Lab for Translational Oncogenomics and Bioinformatics, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Pediatric Precision Oncology Lab, Department of Biomolecular Medicine, Ghent University, 9000 Ghent
| | - Bruno Palhais
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent
| | - Juliette Roels
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent
| | - Malaika Van der Linden
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pathology, Ghent University and Ghent University Hospital, 9000 Ghent
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pathology, Ghent University and Ghent University Hospital, 9000 Ghent
| | - Panagiotis Ntziachristos
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent
| | - Frederik W Van Delft
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne
| | - Marc R Mansour
- Department of Developmental Biology and Cancer, Institute of Child Health, University College London
| | - Tim Pieters
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent
| | - Tim Lammens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium; Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent
| | - Barbara De Moerloose
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, 9000 Ghent
| | - Charles E De Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Kensington, NSW, Australia; School of Clinical Medicine, UNSW Medicine and Health, UNSW Sydney, Sydney, NSW
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Belgium; Unit for Translational Research in Oncology, Department of Diagnostic Sciences, Ghent University, 9000 Ghent.
| | - Pieter Van Vlierberghe
- Lab of Normal and Malignant Hematopoiesis, Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent
| |
Collapse
|
22
|
Wang X, Wang W, Vega F, Quesada AE. Aggressive Mediastinal Lymphomas. Semin Diagn Pathol 2024; 41:125-139. [PMID: 34175178 DOI: 10.1053/j.semdp.2021.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/10/2021] [Indexed: 11/11/2022]
Abstract
The mediastinum contains essentially all major intrathoracic organs except for the lungs. A variety of both benign and malignant tumors can involve the mediastinum, of which lymphoma is the most common malignancy. Compared to secondary mediastinal involvement by systemic lymphomas, primary mediastinal lymphomas are less common with several specific entities that are mainly confined to mediastinal lymph nodes, and/or thymus. This review will summarize the clinical, histologic, immunophenotypic and molecular genetic features of the most common and most aggressive primary mediastinal lymphomas as well as provide suggested immunohistochemistry panels and differential diagnoses.
Collapse
Affiliation(s)
- Xiaoqiong Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Andres E Quesada
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
23
|
Cesana D, Cicalese MP, Calabria A, Merli P, Caruso R, Volpin M, Rudilosso L, Migliavacca M, Barzaghi F, Fossati C, Gazzo F, Pizzi S, Ciolfi A, Bruselles A, Tucci F, Spinozzi G, Pais G, Benedicenti F, Barcella M, Merelli I, Gallina P, Giannelli S, Dionisio F, Scala S, Casiraghi M, Strocchio L, Vinti L, Pacillo L, Draghi E, Cesana M, Riccardo S, Colantuono C, Six E, Cavazzana M, Carlucci F, Schmidt M, Cancrini C, Ciceri F, Vago L, Cacchiarelli D, Gentner B, Naldini L, Tartaglia M, Montini E, Locatelli F, Aiuti A. A case of T-cell acute lymphoblastic leukemia in retroviral gene therapy for ADA-SCID. Nat Commun 2024; 15:3662. [PMID: 38688902 PMCID: PMC11061298 DOI: 10.1038/s41467-024-47866-5] [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: 10/13/2023] [Accepted: 04/10/2024] [Indexed: 05/02/2024] Open
Abstract
Hematopoietic stem cell gene therapy (GT) using a γ-retroviral vector (γ-RV) is an effective treatment for Severe Combined Immunodeficiency due to Adenosine Deaminase deficiency. Here, we describe a case of GT-related T-cell acute lymphoblastic leukemia (T-ALL) that developed 4.7 years after treatment. The patient underwent chemotherapy and haploidentical transplantation and is currently in remission. Blast cells contain a single vector insertion activating the LIM-only protein 2 (LMO2) proto-oncogene, confirmed by physical interaction, and low Adenosine Deaminase (ADA) activity resulting from methylation of viral promoter. The insertion is detected years before T-ALL in multiple lineages, suggesting that further hits occurred in a thymic progenitor. Blast cells contain known and novel somatic mutations as well as germline mutations which may have contributed to transformation. Before T-ALL onset, the insertion profile is similar to those of other ADA-deficient patients. The limited incidence of vector-related adverse events in ADA-deficiency compared to other γ-RV GT trials could be explained by differences in transgenes, background disease and patient's specific factors.
Collapse
Affiliation(s)
- Daniela Cesana
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Cicalese
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Merli
- IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Monica Volpin
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Rudilosso
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Migliavacca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Fossati
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Gazzo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Tucci
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Pais
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Barcella
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Pierangela Gallina
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Giannelli
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Dionisio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Miriam Casiraghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | - Lucia Pacillo
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of Pediatrics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Eleonora Draghi
- Immunogenetics, Leukemia Genomics and Immunobiology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marcella Cesana
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Naples, Italy
| | - Sara Riccardo
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- NEGEDIA S.r.l., Pozzuoli, Italy
| | - Chiara Colantuono
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- NEGEDIA S.r.l., Pozzuoli, Italy
| | - Emmanuelle Six
- Laboratory of Human Lympho-hematopoiesis, INSERM, Paris, France
| | | | - Filippo Carlucci
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Caterina Cancrini
- Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Academic Department of Pediatrics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Department of Systems Medicine University of Rome Tor Vergata, Rome, Italy
| | - Fabio Ciceri
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
- Haematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- Università Vita-Salute San Raffaele, Milan, Italy
- Immunogenetics, Leukemia Genomics and Immunobiology Unit, Division of Immunology, Transplantation and Infectious Diseases, Ospedale San Raffaele Scientific Institute, 20132, Milan, Italy
- Haematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples "Federico II", Naples, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Haematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Paediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
24
|
Suske T, Sorger H, Manhart G, Ruge F, Prutsch N, Zimmerman MW, Eder T, Abdallah DI, Maurer B, Wagner C, Schönefeldt S, Spirk K, Pichler A, Pemovska T, Schweicker C, Pölöske D, Hubanic E, Jungherz D, Müller TA, Aung MMK, Orlova A, Pham HTT, Zimmel K, Krausgruber T, Bock C, Müller M, Dahlhoff M, Boersma A, Rülicke T, Fleck R, de Araujo ED, Gunning PT, Aittokallio T, Mustjoki S, Sanda T, Hartmann S, Grebien F, Hoermann G, Haferlach T, Staber PB, Neubauer HA, Look AT, Herling M, Moriggl R. Hyperactive STAT5 hijacks T cell receptor signaling and drives immature T cell acute lymphoblastic leukemia. J Clin Invest 2024; 134:e168536. [PMID: 38618957 PMCID: PMC11014662 DOI: 10.1172/jci168536] [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: 01/04/2023] [Accepted: 02/27/2024] [Indexed: 04/16/2024] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive immature T cell cancer. Mutations in IL7R have been analyzed genetically, but downstream effector functions such as STAT5A and STAT5B hyperactivation are poorly understood. Here, we studied the most frequent and clinically challenging STAT5BN642H driver in T cell development and immature T cell cancer onset and compared it with STAT5A hyperactive variants in transgenic mice. Enhanced STAT5 activity caused disrupted T cell development and promoted an early T cell progenitor-ALL phenotype, with upregulation of genes involved in T cell receptor (TCR) signaling, even in absence of surface TCR. Importantly, TCR pathway genes were overexpressed in human T-ALL and mature T cell cancers and activation of TCR pathway kinases was STAT5 dependent. We confirmed STAT5 binding to these genes using ChIP-Seq analysis in human T-ALL cells, which were sensitive to pharmacologic inhibition by dual STAT3/5 degraders or ZAP70 tyrosine kinase blockers in vitro and in vivo. We provide genetic and biochemical proof that STAT5A and STAT5B hyperactivation can initiate T-ALL through TCR pathway hijacking and suggest similar mechanisms for other T cell cancers. Thus, STAT5 or TCR component blockade are targeted therapy options, particularly in patients with chemoresistant clones carrying STAT5BN642H.
Collapse
Affiliation(s)
| | | | - Gabriele Manhart
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Frank Ruge
- Institute of Animal Breeding and Genetics and
| | - Nicole Prutsch
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark W. Zimmerman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Eder
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Diaaeldin I. Abdallah
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | - Alexander Pichler
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | - Tea Pemovska
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | - Carmen Schweicker
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | | | | | - Dennis Jungherz
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
| | - Tony Andreas Müller
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
| | | | - Anna Orlova
- Institute of Animal Breeding and Genetics and
| | | | | | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | | | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Auke Boersma
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Rülicke
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Elvin Dominic de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Thomas Gunning
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Janpix, London, United Kingdom
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Satu Mustjoki
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Takaomi Sanda
- Cancer Science Institute of Singapore and Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sylvia Hartmann
- Dr. Senckenberg Institute of Pathology, Goethe University, Frankfurt am Main, Germany
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
- St. Anna Children’s Cancer Research Institute, Vienna, Austria
| | | | | | - Philipp Bernhard Staber
- Department of Medicine I, Clinical Division of Hematology, Medical University of Vienna, Vienna, Austria
| | | | - Alfred Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Marco Herling
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, University of Cologne, Cologne, Germany
- Department of Hematology, Cellular Therapy and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics and
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| |
Collapse
|
25
|
Akkari Y, Baughn LB, Kim A, Karaca E, Raca G, Shao L, Mikhail FM. Section E6.1-6.6 of the American College of Medical Genetics and Genomics (ACMG) Technical Laboratory Standards: Cytogenomic studies of acquired chromosomal abnormalities in neoplastic blood, bone marrow, and lymph nodes. Genet Med 2024; 26:101054. [PMID: 38349293 DOI: 10.1016/j.gim.2023.101054] [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: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 04/09/2024] Open
Abstract
Cytogenomic analyses of acquired clonal chromosomal abnormalities in neoplastic blood, bone marrow, and/or lymph nodes are instrumental in the clinical management of patients with hematologic neoplasms. Cytogenetic analyses assist in the diagnosis of such disorders and can provide important prognostic information. Furthermore, cytogenetic studies can provide crucial information regarding specific genetically defined subtypes of these neoplasms that may have targeted therapies. At time of relapse, cytogenetic analysis can confirm recurrence of the original neoplasm, detect clonal disease evolution, or uncover a new unrelated neoplastic process. This section deals specifically with the technical standards applicable to cytogenomic studies of acquired clonal chromosomal abnormalities in neoplastic blood, bone marrow, and/or lymph nodes. This updated Section E6.1-6.6 supersedes the previous Section E6 in Section E: Clinical Cytogenetics of the American College of Medical Genetics and Genomics Technical Standards for Clinical Genetics Laboratories.
Collapse
Affiliation(s)
- Yassmine Akkari
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH
| | - Linda B Baughn
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Annette Kim
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Ender Karaca
- Department of Pathology, Baylor University Medical Center, Dallas, TX; Texas A&M School of Medicine, Texas A&M University, Dallas, TX
| | - Gordana Raca
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA; Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Lina Shao
- Department of Pathology, University of Michigan, Ann Arbor, MI
| | - Fady M Mikhail
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
26
|
Almeida A, T'Sas S, Pagliaro L, Fijalkowski I, Sleeckx W, Van Steenberge H, Zamponi R, Lintermans B, Van Loocke W, Palhais B, Reekmans A, Bardelli V, Demoen L, Reunes L, Deforce D, Van Nieuwerburgh F, Kentsis A, Ntziachristos P, Van Roy N, De Moerloose B, Mecucci C, La Starza R, Roti G, Goossens S, Van Vlierberghe P, Pieters T. Myb overexpression synergizes with the loss of Pten and is a dependency factor and therapeutic target in T-cell lymphoblastic leukemia. Hemasphere 2024; 8:e51. [PMID: 38463444 PMCID: PMC10924755 DOI: 10.1002/hem3.51] [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: 08/14/2023] [Accepted: 01/28/2024] [Indexed: 03/12/2024] Open
Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that accounts for 10%-15% of pediatric and 25% of adult ALL cases. Although the prognosis of T-ALL has improved over time, the outcome of T-ALL patients with primary resistant or relapsed leukemia remains poor. Therefore, further progress in the treatment of T-ALL requires a better understanding of its biology and the development of more effective precision oncologic therapies. The proto-oncogene MYB is highly expressed in diverse hematologic malignancies, including T-ALLs with genomic aberrations that further potentiate its expression and activity. Previous studies have associated MYB with a malignant role in the pathogenesis of several cancers. However, its role in the induction and maintenance of T-ALL remains relatively poorly understood. In this study, we found that an increased copy number of MYB is associated with higher MYB expression levels, and might be associated with inferior event-free survival of pediatric T-ALL patients. Using our previously described conditional Myb overexpression mice, we generated two distinct MYB-driven T-ALL mouse models. We demonstrated that the overexpression of Myb synergizes with Pten deletion but not with the overexpression of Lmo2 to accelerate the development of T-cell lymphoblastic leukemias. We also showed that MYB is a dependency factor in T-ALL since RNA interference of Myb blocked cell cycle progression and induced apoptosis in both human and murine T-ALL cell lines. Finally, we provide preclinical evidence that targeting the transcriptional activity of MYB can be a useful therapeutic strategy for the treatment of T-ALL.
Collapse
Affiliation(s)
- André Almeida
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Sara T'Sas
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Luca Pagliaro
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Igor Fijalkowski
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Wouter Sleeckx
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Hannah Van Steenberge
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | | | - Béatrice Lintermans
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Wouter Van Loocke
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Bruno Palhais
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Alexandra Reekmans
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Valentina Bardelli
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Lisa Demoen
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Lindy Reunes
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical BiotechnologyGhent UniversityGhentBelgium
| | | | - Alex Kentsis
- Tow Center for Developmental Oncology, Sloan Kettering Institute and Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Panagiotis Ntziachristos
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Nadine Van Roy
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Lab for Translational Oncogenomics and Bioinformatics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Pediatric Precision Oncology Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Department of Pediatric Hematology‐OncologyGhent University HospitalGhentBelgium
| | - Cristina Mecucci
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Roberta La Starza
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Giovanni Roti
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Pieter Van Vlierberghe
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Tim Pieters
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| |
Collapse
|
27
|
Iorgulescu JB, Medeiros LJ, Patel KP. Predictive and prognostic molecular biomarkers in lymphomas. Pathology 2024; 56:239-258. [PMID: 38216400 DOI: 10.1016/j.pathol.2023.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 01/14/2024]
Abstract
Recent advances in molecular diagnostics have markedly expanded our understanding of the genetic underpinnings of lymphomas and catalysed a transformation in not just how we classify lymphomas, but also how we treat, target, and monitor affected patients. Reflecting these advances, the World Health Organization Classification, International Consensus Classification, and National Comprehensive Cancer Network guidelines were recently updated to better integrate these molecular insights into clinical practice. We summarise here the molecular biomarkers of lymphomas with an emphasis on biomarkers that have well-supported prognostic and predictive utility, as well as emerging biomarkers that show promise for clinical practice. These biomarkers include: (1) diagnostic entity-defining genetic abnormalities [e.g., B-cell acute lymphoblastic leukaemia (B-ALL) with KMT2A rearrangement]; (2) molecular alterations that guide patients' prognoses (e.g., TP53 loss frequently conferring worse prognosis); (3) mutations that serve as the targets of, and often a source of acquired resistance to, small molecular inhibitors (e.g., ABL1 tyrosine kinase inhibitors for B-ALL BCR::ABL1, hindered by ABL1 kinase domain resistance mutations); (4) the growing incorporation of molecular measurable residual disease (MRD) in the management of lymphoma patients (e.g., molecular complete response and sequencing MRD-negative criteria in multiple myeloma). Altogether, our review spans the spectrum of lymphoma types, from the genetically defined subclasses of precursor B-cell lymphomas to the highly heterogeneous categories of small and large cell mature B-cell lymphomas, Hodgkin lymphomas, plasma cell neoplasms, and T/NK-cell lymphomas, and provides an expansive summary of our current understanding of their molecular pathology.
Collapse
Affiliation(s)
- J Bryan Iorgulescu
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- Molecular Diagnostics Laboratory, Department of Hematopathology, Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
28
|
Rivera M, Zhang H, Pham J, Isquith J, Zhou QJ, Balaian L, Sasik R, Enlund S, Mark A, Ma W, Holm F, Fisch KM, Kuo DJ, Jamieson C, Jiang Q. Malignant A-to-I RNA editing by ADAR1 drives T cell acute lymphoblastic leukemia relapse via attenuating dsRNA sensing. Cell Rep 2024; 43:113704. [PMID: 38265938 PMCID: PMC10962356 DOI: 10.1016/j.celrep.2024.113704] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/24/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024] Open
Abstract
Leukemia-initiating cells (LICs) are regarded as the origin of leukemia relapse and therapeutic resistance. Identifying direct stemness determinants that fuel LIC self-renewal is critical for developing targeted approaches. Here, we show that the RNA-editing enzyme ADAR1 is a crucial stemness factor that promotes LIC self-renewal by attenuating aberrant double-stranded RNA (dsRNA) sensing. Elevated adenosine-to-inosine editing is a common attribute of relapsed T cell acute lymphoblastic leukemia (T-ALL) regardless of molecular subtype. Consequently, knockdown of ADAR1 severely inhibits LIC self-renewal capacity and prolongs survival in T-ALL patient-derived xenograft models. Mechanistically, ADAR1 directs hyper-editing of immunogenic dsRNA to avoid detection by the innate immune sensor melanoma differentiation-associated protein 5 (MDA5). Moreover, we uncover that the cell-intrinsic level of MDA5 dictates the dependency on the ADAR1-MDA5 axis in T-ALL. Collectively, our results show that ADAR1 functions as a self-renewal factor that limits the sensing of endogenous dsRNA. Thus, targeting ADAR1 presents an effective therapeutic strategy for eliminating T-ALL LICs.
Collapse
Affiliation(s)
- Maria Rivera
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, La Jolla, CA 92037, USA
| | - Haoran Zhang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, La Jolla, CA 92037, USA
| | - Jessica Pham
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jane Isquith
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qingchen Jenny Zhou
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, La Jolla, CA 92037, USA
| | - Larisa Balaian
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Roman Sasik
- Center for Computational Biology & Bioinformatics (CCBB), University of California, San Diego, La Jolla, CA 92093-0681, USA
| | - Sabina Enlund
- Department of Women's and Children's Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, Solna, Sweden
| | - Adam Mark
- Center for Computational Biology & Bioinformatics (CCBB), University of California, San Diego, La Jolla, CA 92093-0681, USA
| | - Wenxue Ma
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Frida Holm
- Department of Women's and Children's Health, Division of Pediatric Oncology and Pediatric Surgery, Karolinska Institutet, Solna, Sweden
| | - Kathleen M Fisch
- Center for Computational Biology & Bioinformatics (CCBB), University of California, San Diego, La Jolla, CA 92093-0681, USA; Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Dennis John Kuo
- Moores Cancer Center, La Jolla, CA 92037, USA; Division of Pediatric Hematology-Oncology, Rady Children's Hospital San Diego, University of California, San Diego, San Diego, CA 92123, USA
| | - Catriona Jamieson
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, La Jolla, CA 92037, USA
| | - Qingfei Jiang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, La Jolla, CA 92037, USA.
| |
Collapse
|
29
|
Zhang J, Duan Y, Wu P, Chang Y, Wang Y, Hu T, Liu C, Chen X, Zong S, Chen X, Wu Y, Jin L, Lan Y, Liu X, Cheng X, Ding F, Li T, Chen X, Guo Y, Chen Y, Yang W, Zhang L, Zou Y, Cheng T, Zhu X, Zhang Y. Clonal evolution dissection reveals that a high MSI2 level promotes chemoresistance in T-cell acute lymphoblastic leukemia. Blood 2024; 143:320-335. [PMID: 37801708 DOI: 10.1182/blood.2023020490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 10/08/2023] Open
Abstract
ABSTRACT T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive cancer with resistant clonal propagation in recurrence. We performed high-throughput droplet-based 5' single-cell RNA with paired T-cell receptor (TCR) sequencing of paired diagnosis-relapse (Dx_Rel) T-ALL samples to dissect the clonal diversities. Two leukemic evolutionary patterns, "clonal shift" and "clonal drift" were unveiled. Targeted single-cell DNA sequencing of paired Dx_Rel T-ALL samples further corroborated the existence of the 2 contrasting clonal evolution patterns, revealing that dynamic transcriptional variation might cause the mutationally static clones to evolve chemotherapy resistance. Analysis of commonly enriched drifted gene signatures showed expression of the RNA-binding protein MSI2 was significantly upregulated in the persistent TCR clonotypes at relapse. Integrated in vitro and in vivo functional studies suggested that MSI2 contributed to the proliferation of T-ALL and promoted chemotherapy resistance through the posttranscriptional regulation of MYC, pinpointing MSI2 as an informative biomarker and novel therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Jingliao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yongjuan Duan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | | | - Yue Wang
- Novogene Co, Ltd, Beijing, China
| | - Tianyuan Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Chao Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoyan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Suyu Zong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoli Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yangping Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Linlin Jin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yang Lan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaoming Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xuelian Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | | | - Tianyu Li
- Wuxi Children's Hospital, Jiangsu, China
| | - Xiaojuan Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ye Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yumei Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenyu Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Li Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yao Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofan Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
30
|
Wei XH, Liu YY. Potential applications of JAK inhibitors, clinically approved drugs against autoimmune diseases, in cancer therapy. Front Pharmacol 2024; 14:1326281. [PMID: 38235120 PMCID: PMC10792058 DOI: 10.3389/fphar.2023.1326281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024] Open
Abstract
Disturbances in immunoregulation may lead to both cancer and autoimmune diseases. Many therapeutic drugs for autoimmune diseases also display anti-tumor efficacy. The Janus kinase/signal transducer and activator of transcription signaling pathways are involved in the secretion of more than 50 distinct cytokines, which have critical roles in inducing autoimmune diseases and tumorigenesis. Thus, Janus kinases have become classical immunotherapeutic targets for immune disease. More than 70 Janus kinase inhibitors have been approved as immunomodulatory drugs for clinical use, of which 12 are used in the treatment of autoimmune diseases. This systematic review aims to elucidate the anti-tumor role of clinically approved Janus kinase inhibitors that were primarily designed for the treatment of autoimmune diseases and their potential for clinical translation as cancer treatments.
Collapse
Affiliation(s)
- Xiao-Huan Wei
- Respiratory and Critical Care Department, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Oncology Department, People’s Hospital of Peixian, Xuzhou, Jiangsu, China
| | - Yuan-Yuan Liu
- Respiratory and Critical Care Department, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
- Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
31
|
Gu A, Schmidt B, Lonsdale A, Jalaldeen R, Kosasih HJ, Brown LM, Sadras T, Ekert PG, Oshlack A. TALLSorts: a T-cell acute lymphoblastic leukemia subtype classifier using RNA-seq expression data. Blood Adv 2023; 7:7402-7406. [PMID: 37903323 PMCID: PMC10758738 DOI: 10.1182/bloodadvances.2023010385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/18/2023] [Accepted: 10/11/2023] [Indexed: 11/01/2023] Open
Affiliation(s)
- Allen Gu
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Breon Schmidt
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Andrew Lonsdale
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Roshan Jalaldeen
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Hansen J. Kosasih
- Children’s Cancer Institute, Kensington, NSW, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | | | - Teresa Sadras
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Paul G. Ekert
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Children’s Cancer Institute, Kensington, NSW, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
- School of Clinical Medicine, University of New South Wales Medicine & Health, University of New South Wales, Kensington, NSW, Australia
| | - Alicia Oshlack
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
32
|
Canarutto D, Asperti C, Vavassori V, Porcellini S, Rovelli E, Paulis M, Ferrari S, Varesi A, Fiumara M, Jacob A, Sergi Sergi L, Visigalli I, Ferrua F, González‐Granado LI, Lougaris V, Finocchi A, Villa A, Radrizzani M, Naldini L. Unbiased assessment of genome integrity and purging of adverse outcomes at the target locus upon editing of CD4 + T-cells for the treatment of Hyper IgM1. EMBO J 2023; 42:e114188. [PMID: 37916874 PMCID: PMC10690452 DOI: 10.15252/embj.2023114188] [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: 04/04/2023] [Revised: 09/15/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Hyper IgM1 is an X-linked combined immunodeficiency caused by CD40LG mutations, potentially treatable with CD4+ T-cell gene editing with Cas9 and a "one-size-fits-most" corrective template. Contrary to established gene therapies, there is limited data on the genomic alterations following long-range gene editing, and no consensus on the relevant assays. We developed drop-off digital PCR assays for unbiased detection of large on-target deletions and found them at high frequency upon editing. Large deletions were also common upon editing different loci and cell types and using alternative Cas9 and template delivery methods. In CD40LG edited T cells, on-target deletions were counter-selected in culture and further purged by enrichment for edited cells using a selector coupled to gene correction. We then validated the sensitivity of optical genome mapping for unbiased detection of genome wide rearrangements and uncovered on-target trapping of one or more vector copies, which do not compromise functionality, upon editing using an integrase defective lentiviral donor template. No other recurring events were detected. Edited patient cells showed faithful reconstitution of CD40LG regulated expression and function with a satisfactory safety profile. Large deletions and donor template integrations should be anticipated and accounted for when designing and testing similar gene editing strategies.
Collapse
Affiliation(s)
- Daniele Canarutto
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
- Università Vita‐Salute San RaffaeleMilanItaly
- Pediatric Immunohematology Unit and BMT ProgramIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Claudia Asperti
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Valentina Vavassori
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Simona Porcellini
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Elisabetta Rovelli
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Marianna Paulis
- Humanitas Clinical and Research Center IRCCSMilanItaly
- UOS Milan UnitIstituto di Ricerca Genetica e Biomedica (IRGB), CNRMilanItaly
| | - Samuele Ferrari
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Angelica Varesi
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Martina Fiumara
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Aurelien Jacob
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Lucia Sergi Sergi
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Ilaria Visigalli
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Francesca Ferrua
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
- Pediatric Immunohematology Unit and BMT ProgramIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Luis Ignacio González‐Granado
- Unidad de Immunodeficiencias Primarias y la Unidad de Hematología y Oncología PediátricaInstituto de Investigacíon Hospital 12 de OctubreMadridSpain
| | | | - Andrea Finocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children's HospitalIstituto di Ricovero e Cura a Carattere ScientificoRomeItaly
| | - Anna Villa
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
- UOS Milan UnitIstituto di Ricerca Genetica e Biomedica (IRGB), CNRMilanItaly
| | - Marina Radrizzani
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene TherapyIRCCS San Raffaele Scientific InstituteMilanItaly
- Università Vita‐Salute San RaffaeleMilanItaly
| |
Collapse
|
33
|
Chen Y, Wu Z, Wang L, Lin M, Jiang P, Wen J, Li J, Hong Y, Zheng X, Yang X, Zheng J, Gale RP, Yang T, Hu J. Targeting nucleolin improves sensitivity to chemotherapy in acute lymphoblastic leukemia. Cell Oncol (Dordr) 2023; 46:1709-1724. [PMID: 37486460 DOI: 10.1007/s13402-023-00837-2] [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] [Accepted: 06/07/2023] [Indexed: 07/25/2023] Open
Abstract
PURPOSE Most patients with acute lymphoblastic leukemia (ALL) are treated with chemotherapy as primary care. Although the treatment response is usually positive, resistance and relapse often occur via unknown mechanisms. The purpose of this study was to identify factors associated with chemotherapy resistance in ALL. Here, we present clinical and experimental evidence that overexpression of nucleolin (NCL), a multifunctional nucleolar protein, is linked to drug resistance in ALL. METHODS NCL mRNA and protein levels were compared between cell lines and patient samples using qRT-PCR and immunoblotting. NCL mRNA levels were compared between patients of different disease stages from our clinic patients' specimens and publicly available ALL patient datasets. Cells and patient-derived xenograft mouse experiments were performed to assess the effect of NCL inhibition on ALL chemotherapy effectiveness. RESULTS Analysis of patient specimens, and publicly available RNA-sequencing datasets revealed a strong correlation between the abundance of NCL and disease relapse or poor survival in B-ALL. Altering NCL expression results in changes in drug sensitivity in ALL cell lines. High levels of NCL upregulated components of the ATP-binding cassette transporters via activation of the ERK pathway, resulting in a decrease in drug accumulation inside the cells. Targeting NCL with AS1411, an NCL-binding oligonucleotide aptamer, significantly increased the sensitivity of ALL cell lines and cells/patient-derived ALL xenograft mice to chemotherapeutic drugs and prolonged mouse survival. CONCLUSION Our results highlight NCL as a prognostic marker in B-ALL and a potential therapeutic target to combat chemotherapy resistance in ALL.
Collapse
Affiliation(s)
- Yanxin Chen
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Zhengjun Wu
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Lingyan Wang
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Minhui Lin
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Peifang Jiang
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Jingjing Wen
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Jiazheng Li
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Yunda Hong
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Xiaoyun Zheng
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Xiaozhu Yang
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Jing Zheng
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Robert Peter Gale
- Haematology Research Centre, Department of Immunology and Inflammation, Imperial college London, South Kensington Campus, London, SW7 2AZ, UK
| | - Ting Yang
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| | - Jianda Hu
- Fujian Provincial Key Laboratory of Hematology, Fujian Institute of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
34
|
Antić Ž, Lentes J, Bergmann AK. Cytogenetics and genomics in pediatric acute lymphoblastic leukaemia. Best Pract Res Clin Haematol 2023; 36:101511. [PMID: 38092485 DOI: 10.1016/j.beha.2023.101511] [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: 03/22/2023] [Revised: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 12/18/2023]
Abstract
The last five decades have witnessed significant improvement in diagnostics, treatment and management of children with acute lymphoblastic leukaemia (ALL). These advancements have become possible through progress in our understanding of the genetic and biological background of ALL, resulting in the introduction of risk-adapted treatment and novel therapeutic targets, e.g., tyrosine kinase inhibitors for BCR::ABL1-positive ALL. Further advances in the taxonomy of ALL and the discovery of new genetic biomarkers and therapeutic targets, as well as the introduction of targeted and immunotherapies into the frontline treatment protocols, may improve management and outcome of children with ALL. In this review we describe the current developments in the (cyto)genetic diagnostics and management of children with ALL, and provide an overview of the most important advances in the genetic classification of ALL. Furthermore, we discuss perspectives resulting from the development of new techniques, including artificial intelligence (AI).
Collapse
Affiliation(s)
- Željko Antić
- Department of Human Genetics, Hannover Medical School (MHH), Hannover, Germany
| | - Jana Lentes
- Department of Human Genetics, Hannover Medical School (MHH), Hannover, Germany
| | - Anke K Bergmann
- Department of Human Genetics, Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
35
|
McCutcheon SR, Swartz AM, Brown MC, Barrera A, McRoberts Amador C, Siklenka K, Humayun L, Ter Weele MA, Isaacs JM, Reddy TE, Allen AS, Nair SK, Antonia SJ, Gersbach CA. Transcriptional and epigenetic regulators of human CD8 + T cell function identified through orthogonal CRISPR screens. Nat Genet 2023; 55:2211-2223. [PMID: 37945901 PMCID: PMC10703699 DOI: 10.1038/s41588-023-01554-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/12/2023]
Abstract
Clinical response to adoptive T cell therapies is associated with the transcriptional and epigenetic state of the cell product. Thus, discovery of regulators of T cell gene networks and their corresponding phenotypes has potential to improve T cell therapies. Here we developed pooled, epigenetic CRISPR screening approaches to systematically profile the effects of activating or repressing 120 transcriptional and epigenetic regulators on human CD8+ T cell state. We found that BATF3 overexpression promoted specific features of memory T cells and attenuated gene programs associated with cytotoxicity, regulatory T cell function, and exhaustion. Upon chronic antigen stimulation, BATF3 overexpression countered phenotypic and epigenetic signatures of T cell exhaustion. Moreover, BATF3 enhanced the potency of CAR T cells in both in vitro and in vivo tumor models and programmed a transcriptional profile that correlates with positive clinical response to adoptive T cell therapy. Finally, we performed CRISPR knockout screens that defined cofactors and downstream mediators of the BATF3 gene network.
Collapse
Affiliation(s)
- Sean R McCutcheon
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Adam M Swartz
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Michael C Brown
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Alejandro Barrera
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Christian McRoberts Amador
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Durham, NC, USA
| | - Keith Siklenka
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Lucas Humayun
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Maria A Ter Weele
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - James M Isaacs
- Duke Cancer Institute Center for Cancer Immunotherapy, Duke University School of Medicine, Durham, NC, USA
| | - Timothy E Reddy
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Andrew S Allen
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Smita K Nair
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
- Duke Cancer Institute Center for Cancer Immunotherapy, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Scott J Antonia
- Duke Cancer Institute Center for Cancer Immunotherapy, Duke University School of Medicine, Durham, NC, USA
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA.
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
36
|
DuVall AS, Wesevich A, Larson RA. Developing Targeted Therapies for T Cell Acute Lymphoblastic Leukemia/Lymphoma. Curr Hematol Malig Rep 2023; 18:217-225. [PMID: 37490229 DOI: 10.1007/s11899-023-00706-7] [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] [Accepted: 07/03/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE OF REVIEW Largely, treatment advances in relapsed and/or refractory acute lymphoblastic leukemia (ALL) have been made in B cell disease leaving T cell ALL reliant upon high-intensity chemotherapy. Recent advances in the understanding of the biology of T-ALL and the improvement in immunotherapies have led to new therapeutic pathways to target and exploit. Here, we review the more promising pathways that are able to be targeted and other therapeutic possibilities for T-ALL. RECENT FINDINGS Preclinical models and early-phase clinical trials have shown promising results in some case in the treatment of T-ALL. Targeting many different pathways could lead to the next advancement in the treatment of relapsed and/or refractory disease. Recent advances in cellular therapies have also shown promise in this space. When reviewing the literature as a whole, targeting important pathways and antigens likely will lead to the next advancement in T-ALL survival since intensifying chemotherapy.
Collapse
Affiliation(s)
- Adam S DuVall
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, USA.
| | - Austin Wesevich
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, USA
| | - Richard A Larson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, USA
| |
Collapse
|
37
|
Paolino J, Dimitrov B, Winger BA, Sandoval-Perez A, Rangarajan AV, Ocasio-Martinez N, Tsai HK, Li Y, Robichaud AL, Khalid D, Hatton C, Gillani R, Polonen P, Dilig A, Gotti G, Kavanagh J, Adhav AA, Gow S, Tsai J, Li YD, Ebert BL, Van Allen EM, Bledsoe J, Kim AS, Tasian SK, Cooper SL, Cooper TM, Hijiya N, Sulis ML, Shukla NN, Magee JA, Mullighan CG, Burke MJ, Luskin MR, Mar BG, Jacobson MP, Harris MH, Stegmaier K, Place AE, Pikman Y. Integration of Genomic Sequencing Drives Therapeutic Targeting of PDGFRA in T-Cell Acute Lymphoblastic Leukemia/Lymphoblastic Lymphoma. Clin Cancer Res 2023; 29:4613-4626. [PMID: 37725576 PMCID: PMC10872648 DOI: 10.1158/1078-0432.ccr-22-2562] [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: 08/16/2022] [Revised: 05/22/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE Patients with relapsed or refractory T-cell acute lymphoblastic leukemia (T-ALL) or lymphoblastic lymphoma (T-LBL) have limited therapeutic options. Clinical use of genomic profiling provides an opportunity to identify targetable alterations to inform therapy. EXPERIMENTAL DESIGN We describe a cohort of 14 pediatric patients with relapsed or refractory T-ALL enrolled on the Leukemia Precision-based Therapy (LEAP) Consortium trial (NCT02670525) and a patient with T-LBL, discovering alterations in platelet-derived growth factor receptor-α (PDGFRA) in 3 of these patients. We identified a novel mutation in PDGFRA, p.D842N, and used an integrated structural modeling and molecular biology approach to characterize mutations at D842 to guide therapeutic targeting. We conducted a preclinical study of avapritinib in a mouse patient-derived xenograft (PDX) model of FIP1L1-PDGFRA and PDGFRA p.D842N leukemia. RESULTS Two patients with T-ALL in the LEAP cohort (14%) had targetable genomic alterations affecting PDGFRA, a FIP1-like 1 protein/PDGFRA (FIP1L1-PDGFRA) fusion and a novel mutation in PDGFRA, p.D842N. The D842N mutation resulted in PDGFRA activation and sensitivity to tested PDGFRA inhibitors. In a T-ALL PDX model, avapritinib treatment led to decreased leukemia burden, significantly prolonged survival, and even cured a subset of mice. Avapritinib treatment was well tolerated and yielded clinical benefit in a patient with refractory T-ALL. CONCLUSIONS Refractory T-ALL has not been fully characterized. Alterations in PDGFRA or other targetable kinases may inform therapy for patients with refractory T-ALL who otherwise have limited treatment options. Clinical genomic profiling, in real time, is needed for fully informed therapeutic decision making.
Collapse
Affiliation(s)
- Jonathan Paolino
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
| | - Boris Dimitrov
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Beth Apsel Winger
- Department of Pediatrics, Division of Hematology/Oncology, Benioff Children’s Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Angelica Sandoval-Perez
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Amith Vikram Rangarajan
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | | | | | - Yuting Li
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Delan Khalid
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Charlie Hatton
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Riaz Gillani
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
| | - Petri Polonen
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | | | - Giacomo Gotti
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Julia Kavanagh
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Asmani A. Adhav
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sean Gow
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jonathan Tsai
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Yen Der Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Jacob Bledsoe
- Department of Pathology, Boston Children’s Hospital, Boston, MA
| | - Annette S. Kim
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Sarah K. Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, and Department of Pediatrics and Abramson Cancer Center at the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Stacy L. Cooper
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD
| | - Todd M. Cooper
- Seattle Children's Hospital, Cancer and Blood Disorders Center, Seattle, WA
| | - Nobuko Hijiya
- Division of Pediatric Hematology/Oncology/Stem Cell Transplantation, Columbia University Irving Medical Center, New York, NY
| | - Maria Luisa Sulis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neerav N. Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jeffrey A. Magee
- Division of Pediatric Hematology/Oncology, Washington University/St. Louis Children's Hospital, St. Louis, MO
| | | | - Michael J. Burke
- Medical College of Wisconsin, Children’s Hospital of Wisconsin, Milwaukee, WI
| | - Marlise R. Luskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | | | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Andrew E. Place
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
| | - Yana Pikman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
38
|
Omari SA, Kosasih HJ, Chung T, de Bock CE. In vitro and in vivo modelling of mutant JAK3/STAT5 signaling in leukemia. Heliyon 2023; 9:e22085. [PMID: 38053908 PMCID: PMC10694137 DOI: 10.1016/j.heliyon.2023.e22085] [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: 10/03/2022] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023] Open
Abstract
Mutations within the IL7-R-JAK-STAT signaling pathway are important drivers of T-cell acute lymphoblastic leukemia (T-ALL). Here we describe the important steps required to generate retroviral particles for the stable expression of mutant JAK3 constructs that induce constitutive JAK/STAT signaling. These are subsequently used for the viral transduction of the IL-3 cytokine-dependent Ba/F3 cell line or murine hematopoietic stem and progenitor cells (HSPCs) for in vitro and in vivo modelling of cytokine-independent growth or leukemia initiation respectively.
Collapse
Affiliation(s)
- Sofia A. Omari
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Hansen J. Kosasih
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Thomas Chung
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| | - Charles E. de Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney 2052, Australia
- School of Clinical Medicine, Medicine and Health, UNSW Sydney, Sydney 2052, Australia
| |
Collapse
|
39
|
Kovach AE, Wood BL. Updates on lymphoblastic leukemia/lymphoma classification and minimal/measurable residual disease analysis. Semin Diagn Pathol 2023; 40:457-471. [PMID: 37953192 DOI: 10.1053/j.semdp.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023]
Abstract
Lymphoblastic leukemia/lymphoma (ALL/LBL), especially certain subtypes, continues to confer morbidity and mortality despite significant therapeutic advances. The pathologic classification of ALL/LBL, especially that of B-ALL, has recently substantially expanded with the identification of several distinct and prognostically important genetic drivers. These discoveries are reflected in both current classification systems, the World Health Organization (WHO) 5th edition and the new International Consensus Classification (ICC). In this article, novel subtypes of B-ALL are reviewed, including DUX4, MEF2D and ZNF384-rearranged B-ALL; the rare pediatric entity B-ALL with TLF3::HLF, now added to the classifications, is discussed; updates to the category of B-ALL with BCR::ABL1-like features (Ph-like B-ALL) are summarized; and emerging genetic subtypes of T-ALL are presented. The second half of the article details current approaches to minimal/measurable residual disease (MRD) detection in B-ALL and T-ALL and presents anticipated challenges to current approaches in the burgeoning era of antigen-directed immunotherapy.
Collapse
Affiliation(s)
- Alexandra E Kovach
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, United States; Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| | - Brent L Wood
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, CA, United States; Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
40
|
Falini B, Lazzi S, Pileri S. A comparison of the International Consensus and 5th WHO classifications of T-cell lymphomas and histiocytic/dendritic cell tumours. Br J Haematol 2023; 203:369-383. [PMID: 37387351 DOI: 10.1111/bjh.18940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/17/2023] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
Since the publication in 2017 of the revised 4th Edition of the World Health Organization (WHO) classification of haematolymphoid tumours, here referred to as WHO-HAEM4, significant clinicopathological, immunophenotypic and molecular advances have been made in the field of lymphomas, contributing to refining the diagnostic criteria of several diseases, upgrading entities previously defined as provisional and identifying new entities. This process has resulted in two recent classification proposals of lymphoid neoplasms: the International Consensus Classification (ICC) and the 5th edition of the WHO classification (WHO-HAEM5). In this paper, we review and compare the two classifications in terms of diagnostic criteria and entity definition, focusing on T-cell lymphomas and histiocytic/dendritic cell tumours. Moreover, we update the genetic data of the various pathological entities. The main goal is to provide a tool to facilitate the work of the pathologists, haematologists and researchers involved in the diagnosis and treatment of these haematological malignancies.
Collapse
Affiliation(s)
- Brunangelo Falini
- Institute of Haematology and CREO, University and Santa Maria della Misericordia Hospital of Perugia, Perugia, Italy
| | - Stefano Lazzi
- Department of Medical Biotechnology, Institute of Pathology, University of Siena, Siena, Italy
| | - Stefano Pileri
- European Institute of Oncology IRCCS, Milan, Italy
- Diatech Pharmacogenetics, Jesi, Italy
| |
Collapse
|
41
|
Chen D, Huang J, Xiao S, Cheng G, Liu Y, Zhao T, Chen C, Yi Y, Peng Y, Cao J. Synthesis, anti-leukemia activity, and molecular docking of novel 3,16-androstenedione derivatives. Steroids 2023; 199:109290. [PMID: 37549776 DOI: 10.1016/j.steroids.2023.109290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023]
Abstract
In this study, we synthesized androsta-4,14-diene-3,16-dione, 12β-hydroxyandrosta-4,14-diene-3,16-dione, and other 3,16-androstenedione derivatives from commercially available dehydroepiandrosterone as a starting material in 9-13 steps with high yields. The bioactivity of the obtained compounds was evaluated. Compounds 14a and 23a were shown to have high antitumor activity against acute lymphoblastic leukemia cell lines Nalm-6 and BALL-1, respectively. Network pharmacology analysis showed that the anti-leukemia activity of compounds 14a and 23a might be related to the JAK2, ABL1 protein, and PI3K/Akt signaling pathways. The molecular docking of compounds 14a and 23a identified possible active sites, with the lowest docking scores for PTGS2 and MAPK14, respectively. In addition, the absorption, distribution, metabolism, and excretion prediction results revealed the drug-likeness of the two compounds. Therefore, compounds 14a and 23a should be considered anti-leukemia candidates in future studies.
Collapse
Affiliation(s)
- Dongjie Chen
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Jiaying Huang
- Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
| | - Shanshan Xiao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Guiguang Cheng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Yaping Liu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Tianrui Zhao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Caixia Chen
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Yongxin Yi
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | - Yungui Peng
- Key Laboratory of Applied Chemistry of Chongqing Municipality, School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China.
| | - Jianxin Cao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
42
|
Higashi T, Yoshida C, Hachiro Y, Nakata C, Takechi A, Yagi T, Miyashita K, Kitada N, Obata R, Hirano T, Hara T, Maki SA. Synthesis and anti-tumor activities in human leukemia-derived cells of polyenylpyrroles with a methyl group at the conjugated polyene terminus. Bioorg Med Chem Lett 2023; 95:129471. [PMID: 37717362 DOI: 10.1016/j.bmcl.2023.129471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
To develop novel drugs for treating T-cell acute lymphoblastic leukemia (T-ALL) and acute myeloid leukemia (AML) which are highly malignant hematological tumors, a series of analogs having a polyenylpyrrole structure of natural compounds (rumbrin and auxarconjugatin B) were synthesized and investigated their structure-activity relationships (SAR) of in vitro anti-T-ALL and anti-AML activities. We obtained three findings: (1) introduction of a methyl group at the conjugated polyene terminus enhanced anti-T-ALL activity, (2) analogs with a 3-chloropyrrole moiety had even higher selectivity for T-ALL cells, and (3) some analogs were effective against AML-derived cells. Among the studied compounds, 3-chloro-2-(8-ethoxycarbonylnona-1,3,5,7-tetraenyl) pyrrole 4e was the most promising candidate of T-ALL- and AML-treating drug. This study provides useful structural information for designing novel drugs treating T-ALL and AML.
Collapse
Affiliation(s)
- Tomoya Higashi
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Chihiro Yoshida
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Yoshifumi Hachiro
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Chihiro Nakata
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Azusa Takechi
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Takuya Yagi
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kazuya Miyashita
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Nobuo Kitada
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Rika Obata
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Takashi Hirano
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Takahiko Hara
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Shojiro A Maki
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan.
| |
Collapse
|
43
|
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
|
44
|
Miyashita K, Yagi T, Kagaya N, Takechi A, Nakata C, Kanda R, Nuriya H, Tanegashima K, Hoyano S, Seki F, Yoshida C, Hachiro Y, Higashi T, Kitada N, Toya T, Kobayashi T, Najima Y, Goyama S, Maki SA, Kitamura T, Doki N, Shin‐ya K, Hara T. Identification of compounds that preferentially suppress the growth of T-cell acute lymphoblastic leukemia-derived cells. Cancer Sci 2023; 114:4032-4040. [PMID: 37522388 PMCID: PMC10551604 DOI: 10.1111/cas.15918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is one of the most frequently occurring cancers in children and is associated with a poor prognosis. Here, we performed large-scale screening of natural compound libraries to identify potential drugs against T-ALL. We identified three low-molecular-weight compounds (auxarconjugatin-B, rumbrin, and lavendamycin) that inhibited the proliferation of the T-ALL cell line CCRF-CEM, but not that of the B lymphoma cell line Raji in a low concentration range. Among them, auxarconjugatin-B and rumbrin commonly contained a polyenyl 3-chloropyrrol in their chemical structure, therefore we chose auxarconjugatin-B for further analyses. Auxarconjugatin-B suppressed the in vitro growth of five human T-ALL cell lines and two T-ALL patient-derived cells, but not that of adult T-cell leukemia patient-derived cells. Cultured normal T cells were several-fold resistant to auxarconjugatin-B. Auxarconjugatin-B and its synthetic analogue Ra#37 depolarized the mitochondrial membrane potential of CCRF-CEM cells within 3 h of treatment. These compounds are promising seeds for developing novel anti-T-ALL drugs.
Collapse
Affiliation(s)
- Kazuya Miyashita
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Division of Cellular TherapyThe Institute of Medical Science, The University of TokyoMinato‐kuJapan
| | - Takuya Yagi
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityBunkyo‐kuJapan
| | - Noritaka Kagaya
- National Institute of Advanced Industrial Science and TechnologyKoto‐kuJapan
| | - Azusa Takechi
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Science, Department of Biological ScienceTokyo Metropolitan UniversityHachioji‐shiJapan
| | - Chihiro Nakata
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityBunkyo‐kuJapan
| | - Risa Kanda
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityBunkyo‐kuJapan
| | - Hideko Nuriya
- Core Technology and Research CenterTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
| | - Kosuke Tanegashima
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
| | - Shota Hoyano
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Science, Department of Biological ScienceTokyo Metropolitan UniversityHachioji‐shiJapan
| | - Fumiya Seki
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityBunkyo‐kuJapan
| | - Chihiro Yoshida
- Department of Engineering Science, Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
- Center for Neuroscience and Biomedical EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Yoshifumi Hachiro
- Department of Engineering Science, Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
- Center for Neuroscience and Biomedical EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Tomoya Higashi
- Department of Engineering Science, Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
- Center for Neuroscience and Biomedical EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Nobuo Kitada
- Department of Engineering Science, Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
- Center for Neuroscience and Biomedical EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Takashi Toya
- Hematology Division, Tokyo Metropolitan Cancer and Infectious diseases CenterKomagome HospitalBunkyo‐kuJapan
| | - Takeshi Kobayashi
- Hematology Division, Tokyo Metropolitan Cancer and Infectious diseases CenterKomagome HospitalBunkyo‐kuJapan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious diseases CenterKomagome HospitalBunkyo‐kuJapan
| | - Susumu Goyama
- Division of Cellular TherapyThe Institute of Medical Science, The University of TokyoMinato‐kuJapan
| | - Shojiro A. Maki
- Department of Engineering Science, Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
- Center for Neuroscience and Biomedical EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Toshio Kitamura
- Division of Cellular TherapyThe Institute of Medical Science, The University of TokyoMinato‐kuJapan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious diseases CenterKomagome HospitalBunkyo‐kuJapan
| | - Kazuo Shin‐ya
- National Institute of Advanced Industrial Science and TechnologyKoto‐kuJapan
| | - Takahiko Hara
- Stem Cell ProjectTokyo Metropolitan Institute of Medical ScienceSetagaya‐kuJapan
- Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityBunkyo‐kuJapan
- Graduate School of Science, Department of Biological ScienceTokyo Metropolitan UniversityHachioji‐shiJapan
| |
Collapse
|
45
|
Tan SH, Tan TK, Yokomori R, Liao M, Huang XZ, Yeoh AEJ, Sanda T. TAL1 hijacks MYCN enhancer that induces MYCN expression and dependence on mevalonate pathway in T-cell acute lymphoblastic leukemia. Leukemia 2023; 37:1969-1981. [PMID: 37591943 DOI: 10.1038/s41375-023-01993-y] [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: 05/18/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
A hallmark of T-cell acute lymphoblastic leukemia (T-ALL) is the dysregulated expression of oncogenic transcription factors (TFs), including TAL1, NOTCH1 and MYC. Rewiring of the transcriptional program disrupts the tightly controlled spatiotemporal expression of downstream target genes, thereby contributing to leukemogenesis. In this study, we first identify an evolutionarily conserved enhancer element controlling the MYCN oncogene (named enhMYCN) that is aberrantly activated by the TAL1 complex in T-ALL cells. TAL1-positive T-ALL cells are highly dependent on MYCN expression for their maintenance in vitro and in xenograft models. Interestingly, MYCN drives the expression of multiple genes involved in the mevalonate pathway, and T-ALL cells are sensitive to inhibition of HMG-CoA reductase (HMGCR), a rate-limiting enzyme of this pathway. Importantly, MYC and MYCN regulate the same targets and compensate for each other. Thus, MYCN-positive T-ALL cells display a dual dependence on the TAL1-MYCN and NOTCH1-MYC pathways. Together, our results demonstrate that enhMYCN-mediated MYCN expression is required for human T-ALL cells and implicate the TAL1-MYCN-HMGCR axis as a potential therapeutic target in T-ALL.
Collapse
Affiliation(s)
- Shi Hao Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Tze King Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Rui Yokomori
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Minghui Liao
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Allen Eng Juh Yeoh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
- Department of Paediatrics, National University of Singapore, Singapore, 119228, Singapore.
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
46
|
Yang A, Luo D, Jia Y, Liu Y, Zhang Z, Li S, Liu R, Zhou J, Wang J. Targeted delivery of AZD5363 to T-cell acute lymphocytic leukemia by mSiO 2-Au nanovehicles. Colloids Surf B Biointerfaces 2023; 230:113505. [PMID: 37574619 DOI: 10.1016/j.colsurfb.2023.113505] [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: 06/05/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023]
Abstract
T-cell acute lymphocytic leukemia (T-ALL) is the most common cancer in children, with a low survival rate because of drug resistance and a high recurrence rate. Targeted delivery of chemotherapy drugs can reduce their side effects and improve their efficacy. The abnormality of phosphatidylinositol-3-kinase/protein kinase B/ mammalian target of rapamycin (PI3K/Akt/mTOR) pathway plays a key role in T-ALL occurrence. AZD5363 is a selective Akt inhibitor with promising therapeutic potential for tumors encoded by the PI3K/Akt/mTOR pathway. However, the toxicity and side effects have limited its application in treating T-ALL. This study aimed to design a delivery system for targeting AZD5363 to T-ALL by sgc8c aptamer designed as mesoporous silica (mSiO2) decorated with Au nanoparticles. The cell-specific targeting and cytotoxicity of mSiO2-Au-AZD5363-Apt were investigated. The mSiO2-Au nanovehicles were found feasible for AZD5363 delivery, with high loading efficiency and pH-responsive release in the acidic lysosome. More importantly, mSiO2-Au-AZD5363-Apt nanovehicles could specifically recognize and enter T-ALL cells in vitro and in vivo, effectively inhibiting the proliferation of CCRF-CEM cells. In conclusion, mSiO2-Au-AZD5363-Apt provided an effective therapeutic method for the targeted treatment of T-ALL.
Collapse
Affiliation(s)
- Aiyun Yang
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Danqing Luo
- Department of Pediatric Hematology Oncology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Yuxuan Jia
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Yuxin Liu
- Department of Chemistry, Capital Normal University, Beijing 100048, China
| | - Zuo Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China
| | - Shen Li
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China
| | - Rong Liu
- Department of Pediatric Hematology Oncology, Children's Hospital, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jing Zhou
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Jianhua Wang
- Translational Medicine Laboratory, Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China.
| |
Collapse
|
47
|
Zhao Y, Jiang S, Tang Y, Zhao L. Venetoclax with CAG regimen for early T-cell precursor acute lymphoblastic leukemia: a case report and literature review. Int J Hematol 2023; 118:483-488. [PMID: 37269505 DOI: 10.1007/s12185-023-03623-w] [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: 02/04/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/05/2023]
Abstract
This article describes a potential treatment for early T-cell precursor acute lymphoblastic leukemia (ETP-ALL), a relatively rare and highly aggressive hematologic malignancy. A 59-year-old woman admitted to our hospital with enlarged cervical lymph nodes, weight loss, abnormal count, and morphology of peripheral blood cells was diagnosed with ETP-ALL according to morphology, immunology, cytogenetics, and molecular biology. The patient initially received two cycles of the VICP regimen, including vincristine, idarubicin, cyclophosphamide, and prednisone, and had a response with positive minimal residual disease (MRD). The patient was then given venetoclax plus the CAG regimen, including aclarubicin, cytosine arabinoside, and granulocyte colony-stimulating factor. After one cycle, the patient achieved complete remission with negative MRD and was eligible for allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Yihan Zhao
- School of the First Clinical Medical, Henan University of Chinese Medicine, Longzihu University Park, Zhengdong New District, 156 Jinshui East Road, Zhengzhou, Henan, China
- Department of Hematology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 200120, China
| | - Shiqing Jiang
- Department of Oncology, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Yujun Tang
- School of the First Clinical Medical, Henan University of Chinese Medicine, Longzihu University Park, Zhengdong New District, 156 Jinshui East Road, Zhengzhou, Henan, China
| | - Lin Zhao
- Department of Hematology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai, 200120, China.
| |
Collapse
|
48
|
Rosenquist R, Bernard E, Erkers T, Scott DW, Itzykson R, Rousselot P, Soulier J, Hutchings M, Östling P, Cavelier L, Fioretos T, Smedby KE. Novel precision medicine approaches and treatment strategies in hematological malignancies. J Intern Med 2023; 294:413-436. [PMID: 37424223 DOI: 10.1111/joim.13697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Genetic testing has been applied for decades in clinical routine diagnostics of hematological malignancies to improve disease (sub)classification, prognostication, patient management, and survival. In recent classifications of hematological malignancies, disease subtypes are defined by key recurrent genetic alterations detected by conventional methods (i.e., cytogenetics, fluorescence in situ hybridization, and targeted sequencing). Hematological malignancies were also one of the first disease areas in which targeted therapies were introduced, the prime example being BCR::ABL1 inhibitors, followed by an increasing number of targeted inhibitors hitting the Achilles' heel of each disease, resulting in a clear patient benefit. Owing to the technical advances in high-throughput sequencing, we can now apply broad genomic tests, including comprehensive gene panels or whole-genome and whole-transcriptome sequencing, to identify clinically important diagnostic, prognostic, and predictive markers. In this review, we give examples of how precision diagnostics has been implemented to guide treatment selection and improve survival in myeloid (myelodysplastic syndromes and acute myeloid leukemia) and lymphoid malignancies (acute lymphoblastic leukemia, diffuse large B-cell lymphoma, and chronic lymphocytic leukemia). We discuss the relevance and potential of monitoring measurable residual disease using ultra-sensitive techniques to assess therapy response and detect early relapses. Finally, we bring up the promising avenue of functional precision medicine, combining ex vivo drug screening with various omics technologies, to provide novel treatment options for patients with advanced disease. Although we are only in the beginning of the field of precision hematology, we foresee rapid development with new types of diagnostics and treatment strategies becoming available to the benefit of our patients.
Collapse
Affiliation(s)
- Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Elsa Bernard
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, USA
- PRISM Center for Personalized Medicine, Gustave Roussy, Villejuif, France
| | - Tom Erkers
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Stockholm, Sweden
| | - David W Scott
- BC Cancer's Centre for Lymphoid Cancer, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Raphael Itzykson
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Rousselot
- Department of Hematology, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Jean Soulier
- Université Paris Cité, Génomes, biologie cellulaire et thérapeutique U944, INSERM, CNRS, Paris, France
- Hématologie Biologique, APHP, Hôpital Saint-Louis, Paris, France
| | - Martin Hutchings
- Department of Haematology and Phase 1 Unit, Rigshospitalet, Copenhagen, Denmark
| | - Päivi Östling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- SciLifeLab, Stockholm, Sweden
| | - Lucia Cavelier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Clinical Genetics, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Thoas Fioretos
- Department of Clinical Genetics, Pathology and Molecular Diagnostics, Office for Medical Services, Region Skåne, Lund, Sweden
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Clinical Genomics Lund, Science for Life Laboratory, Lund University, Lund, Sweden
| | - Karin E Smedby
- Department of Hematology, Karolinska University Hospital, Solna, Stockholm, Sweden
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
49
|
Aktar A, Heit B. Role of the pioneer transcription factor GATA2 in health and disease. J Mol Med (Berl) 2023; 101:1191-1208. [PMID: 37624387 DOI: 10.1007/s00109-023-02359-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023]
Abstract
The transcription factor GATA2 is involved in human diseases ranging from hematopoietic disorders, to cancer, to infectious diseases. GATA2 is one of six GATA-family transcription factors that act as pioneering transcription factors which facilitate the opening of heterochromatin and the subsequent binding of other transcription factors to induce gene expression from previously inaccessible regions of the genome. Although GATA2 is essential for hematopoiesis and lymphangiogenesis, it is also expressed in other tissues such as the lung, prostate gland, gastrointestinal tract, central nervous system, placenta, fetal liver, and fetal heart. Gene or transcriptional abnormalities of GATA2 causes or predisposes patients to several diseases including the hematological cancers acute myeloid leukemia and acute lymphoblastic leukemia, the primary immunodeficiency MonoMAC syndrome, and to cancers of the lung, prostate, uterus, kidney, breast, gastric tract, and ovaries. Recent data has also linked GATA2 expression and mutations to responses to infectious diseases including SARS-CoV-2 and Pneumocystis carinii pneumonia, and to inflammatory disorders such as atherosclerosis. In this article we review the role of GATA2 in the etiology and progression of these various diseases.
Collapse
Affiliation(s)
- Amena Aktar
- Department of Microbiology and Immunology; the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology; the Western Infection, Immunity and Inflammation Centre, The University of Western Ontario, London, ON, N6A 5C1, Canada.
- Robarts Research Institute, London, ON, N6A 3K7, Canada.
| |
Collapse
|
50
|
Jamal M, Lei Y, He H, Zeng X, Bangash HI, Xiao D, Shao L, Zhou F, Zhang Q. CCR9 overexpression promotes T-ALL progression by enhancing cholesterol biosynthesis. Front Pharmacol 2023; 14:1257289. [PMID: 37745085 PMCID: PMC10512069 DOI: 10.3389/fphar.2023.1257289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy of the lymphoid progenitor cells, contributing to ∼ 20% of the total ALL cases, with a higher prevalence in adults than children. Despite the important role of human T-ALL cell lines in understanding the pathobiology of the disease, a detailed comparison of the tumorigenic potentials of two commonly used T-ALL cell lines, MOLT4 and JURKAT cells, is still lacking. Methodology: In the present study, NOD-Prkdc scid IL2rgd ull (NTG) mice were intravenously injected with MOLT4, JURKAT cells, and PBS as a control. The leukemiac cell homing/infiltration into the bone marrow, blood, liver and spleen was investigated for bioluminescence imaging, flow cytometry, and immunohistochemistry staining. Gene expression profiling of the two cell lines was performed via RNA-seq to identify the differentially expressed genes (DEGs). CCR9 identified as a DEG, was further screened for its role in invasion and metastasis in both cell lines in vitro. Moreover, a JURKAT cell line with overexpressed CCR9 (Jurkat-OeCCR9) was investigated for T-ALL formation in the NTG mice as compared to the GFP control. Jurkat-OeCCR9 cells were then subjected to transcriptome analysis to identify the genes and pathways associated with the upregulation of CCR9 leading to enhanced tumirogenesis. The DEGs of the CCR9-associated upregulation were validated both at mRNA and protein levels. Simvastatin was used to assess the effect of cholesterol biosynthesis inhibition on the aggressiveness of T-ALL cells. Results: Comparison of the leukemogenic potentials of the two T-ALL cell lines showed the relatively higher leukemogenic potential of MOLT4 cells, characterized by their enhanced tissue infiltration in NOD-PrkdcscidIL2rgdull (NTG) mice. Transcriptmoe analysis of the two cell lines revealed numerous DEGs, including CCR9, enriched in vital signaling pathways associated with growth and proliferation. Notably, the upregulation of CCR9 also promoted the tissue infiltration of JURKAT cells in vitro and in NTG mice. Transcriptome analysis revealed that CCR9 overexpression facilitated cholesterol production by upregulating the expression of the transcriptional factor SREBF2, and the downstream genes: MSMO1, MVD, HMGCS1, and HMGCR, which was then corroborated at the protein levels. Notably, simvastatin treatment reduced the migration of the CCR9-overexpressing JURKAT cells, suggesting the importance of cholesterol in T-ALL progression. Conclusions: This study highlights the distinct tumorigenic potentials of two T-ALL cell lines and reveals CCR9-regulated enhanced cholesterol biosynthesis in T-ALL.
Collapse
Affiliation(s)
- Muhammad Jamal
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yufei Lei
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hengjing He
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xingruo Zeng
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hina Iqbal Bangash
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Di Xiao
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Quiping Zhang
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan University, Wuhan, China
| |
Collapse
|