1
|
Catarino TA, Pacheco-Leyva I, Baessa M, Pereira JL, R Dos Santos N. Transgenic αβ TCR tonic signaling is leukemogenic while strong stimulation is leukemia suppressive. J Leukoc Biol 2025; 117:qiae249. [PMID: 39574201 DOI: 10.1093/jleuko/qiae249] [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: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 03/30/2025] Open
Abstract
The pre-T cell receptor (TCR) and TCR complexes are frequently expressed in T cell acute lymphoblastic leukemia (T-ALL), an aggressive T cell precursor malignancy. Although mutations in TCR components are infrequent in T-ALL, earlier research indicated that transgenic αβ TCR expression in mouse T cell precursors promoted T-ALL development. However, we recently found that stimulation of TCR signaling in T-ALL induced leukemic cell apoptosis and suppressed leukemia. Our aim was to elucidate if a given αβ TCR complex has a dual role in leukemogenesis depending on the nature of the stimulus. We demonstrate that transgenic expression of the Marilyn αβ TCR, specific for the H-Y male antigen presented by major histocompatibility complex class II, triggers T-ALL development exclusively in female mice. This T-ALL exhibited Notch1 mutations, Cdkn2a copy number loss, and immature immunophenotype, and infiltrated both lymphoid and nonlymphoid organs. Furthermore, leukemic cells expressed surface CD5, a marker of tonic TCR signaling. T-ALL efficiently developed in Rag2-deficient Marilyn transgenic females, indicating that Rag2-mediated recombination is not implicated in this T-ALL model. T-ALL development was also observed in the OT-I TCR transgenic mouse model, but it did not occur when major histocompatibility complex class I was abrogated through genetic inactivation of β2-microglobulin. Remarkably, exposure of Marilyn female T-ALL cells to endogenous agonist antigens in male recipient mice or exogenous peptides in female recipient mice resulted in T-ALL apoptosis and prolonged mouse survival. These findings underscore the dual role of the same αβ TCR complex in T-ALL, in which tonic stimulation is leukemogenic, while strong stimulation suppresses leukemia.
Collapse
MESH Headings
- Animals
- Mice, Transgenic
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Female
- Signal Transduction/immunology
- Mice
- Male
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology
- Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics
- DNA-Binding Proteins/genetics
- H-Y Antigen/immunology
- Mice, Inbred C57BL
Collapse
Affiliation(s)
- Telmo A Catarino
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology (IPATIMUP), Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ivette Pacheco-Leyva
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology (IPATIMUP), Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Marina Baessa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology (IPATIMUP), Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - João L Pereira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology (IPATIMUP), Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
| | - Nuno R Dos Santos
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Institute of Molecular Pathology and Immunology (IPATIMUP), Universidade do Porto, Rua Júlio Amaral de Carvalho, 45, 4200-135 Porto, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
2
|
Cwynarski K, Iacoboni G, Tholouli E, Menne T, Irvine DA, Balasubramaniam N, Wood L, Shang J, Xue E, Zhang Y, Basilico S, Neves M, Raymond M, Scott I, El-Kholy M, Jha R, Dainton-Smith H, Hussain R, Day W, Ferrari M, Thomas S, Lilova K, Brugger W, Marafioti T, Lao-Sirieix P, Maciocia P, Pule M. TRBC1-CAR T cell therapy in peripheral T cell lymphoma: a phase 1/2 trial. Nat Med 2025; 31:137-143. [PMID: 39528665 PMCID: PMC11750712 DOI: 10.1038/s41591-024-03326-7] [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: 01/18/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Relapsed/refractory peripheral T cell lymphomas (PTCLs) are aggressive tumors with a poor prognosis. Unlike B cell lymphomas, treatment of PTCL has not benefited from advances in immunotherapy. This is largely due to a lack of suitable target antigens that discriminate malignant from normal T cells, thus avoiding severe immunosuppression consequent to depletion of the entire T cell compartment. We recently described a targeting strategy based on the mutually exclusive expression of T cell antigen receptor beta-chain constant domain (TRBC) 1 and 2. Selective targeting of the T cell antigen receptor beta-chain expressed by the (clonal) malignancy spares normal T cells expressing the other chain. The LibraT1 study is an ongoing, multicenter, international, single-arm phase 1/2 study of TRBC1-directed autologous chimeric antigen receptor (CAR) T cells (AUTO4) in relapsed/refractory TRBC1-positive PTCL. Primary objectives were assessment of safety and tolerability of AUTO4 infusion. Key secondary endpoints included efficacy, CAR T cell expansion and persistence. Here we describe the findings from dose escalation in LibraT1 in the first ten patients, in a non-prespecified interim analysis. AUTO4 resulted in low frequency of severe immunotoxicity, with one of ten patients developing grade 3 cytokine release syndrome. Complete metabolic response was observed in four of ten evaluable patients, with remissions being durable beyond 1 year in two patients. While an absence of circulating CAR T cells was observed, CAR T cells were readily detected in lymph node biopsy samples from sites of original disease suggesting homing to tumor sites. These results support the continuing exploration of TRBC1 targeting in PTCL. ClinicalTrials.gov registration: NCT03590574 .
Collapse
MESH Headings
- Humans
- Lymphoma, T-Cell, Peripheral/therapy
- Lymphoma, T-Cell, Peripheral/immunology
- Lymphoma, T-Cell, Peripheral/pathology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Male
- Female
- Middle Aged
- Adult
- Aged
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Kate Cwynarski
- Department of Haematology, University College London, London, UK
- University College London Hospital, London, UK
| | - Gloria Iacoboni
- Department of Haematology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Eleni Tholouli
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| | - Tobias Menne
- The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | | | - Leigh Wood
- University College London Hospital, London, UK
| | | | - Eric Xue
- Autolus Therapeutics, London, UK
| | | | | | | | | | | | | | - Ram Jha
- Autolus Therapeutics, London, UK
| | | | | | | | | | | | | | | | | | | | - Paul Maciocia
- Department of Haematology, University College London, London, UK
- University College London Hospital, London, UK
| | - Martin Pule
- Department of Haematology, University College London, London, UK.
- Autolus Therapeutics, London, UK.
| |
Collapse
|
3
|
Ávila Ávila A, Nuantang K, Oliveira ML, Druillennec S, Zaniboni B, Lengliné E, Asnafi V, Ghysdael J, Tran Quang C. Targeting the TNF/IAP pathway synergizes with anti-CD3 immunotherapy in T-cell acute lymphoblastic leukemia. Blood 2024; 143:2166-2177. [PMID: 38437728 PMCID: PMC11143533 DOI: 10.1182/blood.2023022455] [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: 09/07/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
ABSTRACT T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy. Current treatments, based on intensive chemotherapy regimens provide overall survival rates of ∼85% in children and <50% in adults, calling the search of new therapeutic options. We previously reported that targeting the T-cell receptor (TCR) in T-ALL with anti-CD3 (αCD3) monoclonal antibodies (mAbs) enforces a molecular program akin to thymic negative selection, a major developmental checkpoint in normal T-cell development; induces leukemic cell death; and impairs leukemia progression to ultimately improve host survival. However, αCD3 monotherapy resulted in relapse. To find out actionable targets able to re-enforce leukemic cells' vulnerability to αCD3 mAbs, including the clinically relevant teplizumab, we identified the molecular program induced by αCD3 mAbs in patient-derived xenografts derived from T-ALL cases. Using large-scale transcriptomic analysis, we found prominent expression of tumor necrosis factor α (TNFα), lymphotoxin α (LTα), and multiple components of the "TNFα via NF-κB signaling" pathway in anti-CD3-treated T-ALL. We show in vivo that etanercept, a sink for TNFα/LTα, enhances αCD3 antileukemic properties, indicating that TNF/TNF receptor (TNFR) survival pathways interferes with TCR-induced leukemic cell death. However, suppression of TNF-mediated survival and switch to TNFR-mediated cell death through inhibition of cellular inhibitor of apoptosis protein-1/2 (cIAP1/2) with the second mitochondrial-derived activator of caspases (SMAC) mimetic birinapant synergizes with αCD3 to impair leukemia expansion in a receptor-interacting serine/threonine-protein kinase 1-dependent manner and improve mice survival. Thus, our results advocate the use of either TNFα/LTα inhibitors, or birinapant/other SMAC mimetics to improve anti-CD3 immunotherapy in T-ALL.
Collapse
Affiliation(s)
- Andrea Ávila Ávila
- Institut Curie, Orsay, France
- Centre National de la Recherche Scientifique UMR3348, Centre Universitaire, Orsay, France
- INSERM, U1278, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Kanokporn Nuantang
- Institut Curie, Orsay, France
- Centre National de la Recherche Scientifique UMR3348, Centre Universitaire, Orsay, France
- INSERM, U1278, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Mariana L. Oliveira
- Institut Curie, Orsay, France
- Centre National de la Recherche Scientifique UMR3348, Centre Universitaire, Orsay, France
- INSERM, U1278, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Sabine Druillennec
- Institut Curie, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
- INSERM, U1021, Centre Universitaire, Orsay, France
| | - Benedetta Zaniboni
- Institut Curie, Orsay, France
- Centre National de la Recherche Scientifique UMR3348, Centre Universitaire, Orsay, France
- INSERM, U1278, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Etienne Lengliné
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint Louis, Unité d’Hématologie, Paris, France
| | - Vahid Asnafi
- Université Paris Cité, Institut Necker-Enfants Malades, INSERM U1151, Paris, France
| | - Jacques Ghysdael
- Institut Curie, Orsay, France
- Centre National de la Recherche Scientifique UMR3348, Centre Universitaire, Orsay, France
- INSERM, U1278, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Christine Tran Quang
- Institut Curie, Orsay, France
- Centre National de la Recherche Scientifique UMR3348, Centre Universitaire, Orsay, France
- INSERM, U1278, Orsay, France
- University Paris Sud, Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| |
Collapse
|
4
|
Sawyer EM, Kraft AS. SMACing down relapsed T-ALL. Blood 2024; 143:2116-2117. [PMID: 38780917 DOI: 10.1182/blood.2024024304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Affiliation(s)
| | - Andrew S Kraft
- Vortex Biotechnology Corporation
- University of Colorado Anschutz Medical Center
| |
Collapse
|
5
|
Carty SA, Murga-Zamalloa CA, Wilcox RA. SOHO State of the Art Updates and Next Questions | New Pathways and New Targets in PTCL: Staying on Target. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:561-574. [PMID: 37142534 PMCID: PMC10565700 DOI: 10.1016/j.clml.2023.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/05/2023] [Accepted: 04/16/2023] [Indexed: 05/06/2023]
Abstract
While the peripheral T-cell lymphomas (PTCL) remain a therapeutic challenge, and increasingly account for a disproportionate number of lymphoma-related deaths, improved understanding of disease pathogenesis and classification, and the development of novel therapeutic agents over the past decade, all provide reasons for a more optimistic outlook in the next. Despite their genetic and molecular heterogeneity, many PTCL are dependent upon signaling input provided by antigen, costimulatory, and cytokine receptors. While gain-of-function alterations effecting these pathways are recurrently observed in many PTCL, more often than not, signaling remains ligand-and tumor microenvironment (TME)-dependent. Consequently, the TME and its constituents are increasingly recognized as "on target". Utilizing a "3 signal" model, we will review new-and old-therapeutic targets that are relevant for the more common nodal PTCL subtypes.
Collapse
Affiliation(s)
- Shannon A Carty
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | | | - Ryan A Wilcox
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
6
|
Caracciolo D, Mancuso A, Polerà N, Froio C, D'Aquino G, Riillo C, Tagliaferri P, Tassone P. The emerging scenario of immunotherapy for T-cell Acute Lymphoblastic Leukemia: advances, challenges and future perspectives. Exp Hematol Oncol 2023; 12:5. [PMID: 36624522 PMCID: PMC9828428 DOI: 10.1186/s40164-022-00368-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a challenging pediatric and adult haematologic disease still associated with an unsatisfactory cure rate. Unlike B-ALL, the availability of novel therapeutic options to definitively improve the life expectancy for relapsed/resistant patients is poor. Indeed, the shared expression of surface targets among normal and neoplastic T-cells still limits the efficacy and may induce fratricide effects, hampering the use of innovative immunotherapeutic strategies. However, novel monoclonal antibodies, bispecific T-cell engagers (BTCEs), and chimeric antigen receptors (CAR) T-cells recently showed encouraging results and some of them are in an advanced stage of pre-clinical development or are currently under investigation in clinical trials. Here, we review this exciting scenario focusing on most relevant advances, challenges, and perspectives of the emerging landscape of immunotherapy of T-cell malignancies.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Antonia Mancuso
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Caterina Froio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Giuseppe D'Aquino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy
| | | | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Catanzaro, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Xu X, Zhang W, Xuan L, Yu Y, Zheng W, Tao F, Nemechek J, He C, Ma W, Han X, Xie S, Zhao M, Wang J, Qu Y, Liu Q, Perry JM, Jiang L, Zhao M. PD-1 signalling defines and protects leukaemic stem cells from T cell receptor-induced cell death in T cell acute lymphoblastic leukaemia. Nat Cell Biol 2023; 25:170-182. [PMID: 36624186 DOI: 10.1038/s41556-022-01050-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 11/10/2022] [Indexed: 01/11/2023]
Abstract
T cell acute lymphoblastic leukaemia (T-ALL) is an aggressive malignancy with poor prognosis, but a decisive marker and effective treatment for leukaemia stem cells (LSCs) remain unclear. Here, using lineage tracing, limiting dilution assays and in vivo live imaging approaches, we identify rare inhibitory receptor programmed cell death 1 (PD-1)-expressing cells that reside at the apex of leukaemia hierarchy for initiation and relapse in T-ALL. Ablation of PD-1-expressing cells, deletion of PD-1 in T-ALL cells or blockade of PD-1 or PD-1 ligand 1 significantly eradicated LSCs and suppressed disease progression. Combination therapy using PD-1 blockade and chemotherapy substantially extended the survival of mice engrafted with mouse or human T-ALL cells. Mechanistically, PD-1+ LSCs had high NOTCH1-MYC activity for disease initiation. Furthermore, PD-1 signalling maintained quiescence and protected LSCs against T cell receptor-signal-induced apoptosis. Overall, our data highlight the hierarchy of leukaemia by identifying PD-1+ LSCs and provide a therapeutic approach for the elimination of LSCs through PD-1 blockade in T-ALL.
Collapse
Affiliation(s)
- Xi Xu
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenwen Zhang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanhui Yu
- Department of Hematology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, China
| | - Wen Zheng
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Fang Tao
- Children's Mercy Kansas City, Kansas City, MO, USA
| | | | - Chong He
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Ma
- Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xue Han
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Siyu Xie
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Minyi Zhao
- Department of Hematology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Wang
- Department of Pediatric Hematology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuhua Qu
- Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - John M Perry
- Children's Mercy Kansas City, Kansas City, MO, USA.,University of Kansas Medical Center, Kansas City, KS, USA.,University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Linjia Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Meng Zhao
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory of Stem Cells and Tissue Engineering (Ministry of Education), Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
8
|
Laukkanen S, Veloso A, Yan C, Oksa L, Alpert EJ, Do D, Hyvärinen N, McCarthy K, Adhikari A, Yang Q, Iyer S, Garcia SP, Pello A, Ruokoranta T, Moisio S, Adhikari S, Yoder JA, Gallagher K, Whelton L, Allen JR, Jin AH, Loontiens S, Heinäniemi M, Kelliher M, Heckman CA, Lohi O, Langenau DM. Therapeutic targeting of LCK tyrosine kinase and mTOR signaling in T-cell acute lymphoblastic leukemia. Blood 2022; 140:1891-1906. [PMID: 35544598 PMCID: PMC10082361 DOI: 10.1182/blood.2021015106] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
Relapse and refractory T-cell acute lymphoblastic leukemia (T-ALL) has a poor prognosis, and new combination therapies are sorely needed. Here, we used an ex vivo high-throughput screening platform to identify drug combinations that kill zebrafish T-ALL and then validated top drug combinations for preclinical efficacy in human disease. This work uncovered potent drug synergies between AKT/mTORC1 (mammalian target of rapamycin complex 1) inhibitors and the general tyrosine kinase inhibitor dasatinib. Importantly, these same drug combinations effectively killed a subset of relapse and dexamethasone-resistant zebrafish T-ALL. Clinical trials are currently underway using the combination of mTORC1 inhibitor temsirolimus and dasatinib in other pediatric cancer indications, leading us to prioritize this therapy for preclinical testing. This combination effectively curbed T-ALL growth in human cell lines and primary human T-ALL and was well tolerated and effective in suppressing leukemia growth in patient-derived xenografts (PDX) grown in mice. Mechanistically, dasatinib inhibited phosphorylation and activation of the lymphocyte-specific protein tyrosine kinase (LCK) to blunt the T-cell receptor (TCR) signaling pathway, and when complexed with mTORC1 inhibition, induced potent T-ALL cell killing through reducing MCL-1 protein expression. In total, our work uncovered unexpected roles for the LCK kinase and its regulation of downstream TCR signaling in suppressing apoptosis and driving continued leukemia growth. Analysis of a wide array of primary human T-ALLs and PDXs grown in mice suggest that combination of temsirolimus and dasatinib treatment will be efficacious for a large fraction of human T-ALLs.
Collapse
Affiliation(s)
- Saara Laukkanen
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alexandra Veloso
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Chuan Yan
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Laura Oksa
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Eric J. Alpert
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Daniel Do
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Noora Hyvärinen
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Karin McCarthy
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Abhinav Adhikari
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Qiqi Yang
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Sowmya Iyer
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Sara P. Garcia
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Annukka Pello
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Tanja Ruokoranta
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sanni Moisio
- The Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sadiksha Adhikari
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Jeffrey A. Yoder
- Department of Molecular Biomedical Sciences, Comparative Medicine Institute, and Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| | - Kayleigh Gallagher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Lauren Whelton
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - James R. Allen
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Alex H. Jin
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| | - Siebe Loontiens
- Cancer Research Institute Ghent and Center for Medical Genetics, Ghent, Belgium
| | - Merja Heinäniemi
- The Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Michelle Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Olli Lohi
- Tampere Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere University Hospital, Tays Cancer Center, Tampere, Finland
| | - David M. Langenau
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA
- Harvard Stem Cell Institute, Boston, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
9
|
Antoszewski M, Fournier N, Ruiz Buendía GA, Lourenco J, Liu Y, Sugrue T, Dubey C, Nkosi M, Pritchard CE, Huijbers IJ, Segat GC, Alonso-Moreno S, Serracanta E, Belver L, Ferrando AA, Ciriello G, Weng AP, Koch U, Radtke F. Tcf1 is essential for initiation of oncogenic Notch1-driven chromatin topology in T-ALL. Blood 2022; 139:2483-2498. [PMID: 35020836 PMCID: PMC9710489 DOI: 10.1182/blood.2021012077] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/22/2021] [Indexed: 01/16/2023] Open
Abstract
NOTCH1 is a well-established lineage specifier for T cells and among the most frequently mutated genes throughout all subclasses of T cell acute lymphoblastic leukemia (T-ALL). How oncogenic NOTCH1 signaling launches a leukemia-prone chromatin landscape during T-ALL initiation is unknown. Here we demonstrate an essential role for the high-mobility-group transcription factor Tcf1 in orchestrating chromatin accessibility and topology, allowing aberrant Notch1 signaling to convey its oncogenic function. Although essential, Tcf1 is not sufficient to initiate leukemia. The formation of a leukemia-prone epigenetic landscape at the distal Notch1-regulated Myc enhancer, which is fundamental to this disease, is Tcf1-dependent and occurs within the earliest progenitor stage even before cells adopt a T lymphocyte or leukemic fate. Moreover, we discovered a unique evolutionarily conserved Tcf1-regulated enhancer element in the distal Myc-enhancer, which is important for the transition of preleukemic cells to full-blown disease.
Collapse
Affiliation(s)
- Mateusz Antoszewski
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Nadine Fournier
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Gustavo A. Ruiz Buendía
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Joao Lourenco
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Yuanlong Liu
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Tara Sugrue
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Botnar Research Centre for Child Health, University of Basel & ETH Zürich, Basel, Switzerland
| | - Christelle Dubey
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- INSELSPITAL, Universitätsspital Bern, Universitätsklinik für Thoraxchirurgie, Forschungsabteilung Thoraxchirurgie, Bern, Switzerland
| | - Marianne Nkosi
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Colin E.J. Pritchard
- Mouse Clinic for Cancer & Aging (MCCA)/Transgenic Core Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ivo J. Huijbers
- Mouse Clinic for Cancer & Aging (MCCA)/Transgenic Core Facility, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Laura Belver
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Catalan Institute of Oncology-Immuno Procure, Barcelona, Spain
| | - Adolfo A. Ferrando
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY
| | - Giovanni Ciriello
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne (UNIL), Lausanne, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Andrew P. Weng
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada
| | - Ute Koch
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| | - Freddy Radtke
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Sciences, Swiss Institute for Experimental Cancer Research (ISREC), Lausanne, Switzerland
- Swiss Cancer Center Leman (SCCL), Lausanne, Switzerland
| |
Collapse
|
10
|
Shah K, Al-Haidari A, Sun J, Kazi JU. T cell receptor (TCR) signaling in health and disease. Signal Transduct Target Ther 2021; 6:412. [PMID: 34897277 PMCID: PMC8666445 DOI: 10.1038/s41392-021-00823-w] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 11/02/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Interaction of the T cell receptor (TCR) with an MHC-antigenic peptide complex results in changes at the molecular and cellular levels in T cells. The outside environmental cues are translated into various signal transduction pathways within the cell, which mediate the activation of various genes with the help of specific transcription factors. These signaling networks propagate with the help of various effector enzymes, such as kinases, phosphatases, and phospholipases. Integration of these disparate signal transduction pathways is done with the help of adaptor proteins that are non-enzymatic in function and that serve as a scaffold for various protein-protein interactions. This process aids in connecting the proximal to distal signaling pathways, thereby contributing to the full activation of T cells. This review provides a comprehensive snapshot of the various molecules involved in regulating T cell receptor signaling, covering both enzymes and adaptors, and will discuss their role in human disease.
Collapse
Affiliation(s)
- Kinjal Shah
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Amr Al-Haidari
- Clinical Genetics and Pathology, Skåne University Hospital, Region Skåne, Lund, Sweden
- Clinical Sciences Department, Surgery Research Unit, Lund University, Malmö, Sweden
| | - Jianmin Sun
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Science and Technology center, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Julhash U Kazi
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
- Lund Stem Cell Center, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
11
|
Andrieu GP, Kohn M, Simonin M, Smith CL, Cieslak A, Dourthe MÉ, Charbonnier G, Graux C, Huguet F, Lhéritier V, Dombret H, Spicuglia S, Rousselot P, Boissel N, Asnafi V. PRC2 loss of function confers a targetable vulnerability to BET proteins in T-ALL. Blood 2021; 138:1855-1869. [PMID: 34125178 PMCID: PMC9642784 DOI: 10.1182/blood.2020010081] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/21/2021] [Indexed: 11/20/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a group of aggressive hematological cancers with dismal outcomes that are in need of new therapeutic options. Polycomb repressor complex 2 (PRC2) loss-of-function alterations were reported in pediatric T-ALL, yet their clinical relevance and functional consequences remain elusive. Here, we extensively analyzed PRC2 alterations in a large series of 218 adult T-ALL patients. We found that PRC2 genetic lesions are frequent events in T-ALL and are not restricted to early thymic precursor ALL. PRC2 loss of function associates with activating mutations of the IL7R/JAK/STAT pathway. PRC2-altered T-ALL patients respond poorly to prednisone and have low bone marrow blast clearance and persistent minimal residual disease. Furthermore, we identified that PRC2 loss of function profoundly reshapes the genetic and epigenetic landscapes, leading to the reactivation of stem cell programs that cooperate with bromodomain and extraterminal (BET) proteins to sustain T-ALL. This study identifies BET proteins as key mediators of the PRC2 loss of function-induced remodeling. Our data have uncovered a targetable vulnerability to BET inhibition that can be exploited to treat PRC2-altered T-ALL patients.
Collapse
Affiliation(s)
- Guillaume P Andrieu
- Institut Necker Enfants-Malades, Team 2, INSERM Unité1151, Paris, France
- Hôpital Necker Enfants-Malades, Laboratoire d'Onco-Hématologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université de Paris, Paris, France
| | - Milena Kohn
- Hôpital Necker Enfants-Malades, Laboratoire d'Onco-Hématologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Mathieu Simonin
- Institut Necker Enfants-Malades, Team 2, INSERM Unité1151, Paris, France
- Hôpital Necker Enfants-Malades, Laboratoire d'Onco-Hématologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université de Paris, Paris, France
| | - Charlotte L Smith
- Institut Necker Enfants-Malades, Team 2, INSERM Unité1151, Paris, France
- Université de Paris, Paris, France
| | - Agata Cieslak
- Institut Necker Enfants-Malades, Team 2, INSERM Unité1151, Paris, France
- Hôpital Necker Enfants-Malades, Laboratoire d'Onco-Hématologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Marie-Émilie Dourthe
- Institut Necker Enfants-Malades, Team 2, INSERM Unité1151, Paris, France
- Hôpital Necker Enfants-Malades, Laboratoire d'Onco-Hématologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université de Paris, Paris, France
| | - Guillaume Charbonnier
- Aix-Marseille University, Theories and Approaches of Genomic Complexity (TAGC), INSERM Unité Mixte de Recherche (UMR)1090 13288 Marseille, France
| | - Carlos Graux
- Université Catholique de Louvain, Centre Hospitalier Universitaire UCLouvaine Namur-Godinne, Service d'Hématologie, Yvoir, Belgium
| | | | | | - Hervé Dombret
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - Salvatore Spicuglia
- Aix-Marseille University, Theories and Approaches of Genomic Complexity (TAGC), INSERM Unité Mixte de Recherche (UMR)1090 13288 Marseille, France
| | - Philippe Rousselot
- Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, 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
- Institut Necker Enfants-Malades, Team 2, INSERM Unité1151, Paris, France
- Hôpital Necker Enfants-Malades, Laboratoire d'Onco-Hématologie, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université de Paris, Paris, France
| |
Collapse
|
12
|
Silva A, Almeida ARM, Cachucho A, Neto JL, Demeyer S, de Matos M, Hogan T, Li Y, Meijerink J, Cools J, Grosso AR, Seddon B, Barata JT. Overexpression of wild-type IL-7Rα promotes T-cell acute lymphoblastic leukemia/lymphoma. Blood 2021; 138:1040-1052. [PMID: 33970999 PMCID: PMC8462360 DOI: 10.1182/blood.2019000553] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/15/2021] [Indexed: 12/02/2022] Open
Abstract
Tight regulation of IL-7Rα expression is essential for normal T-cell development. IL-7Rα gain-of-function mutations are known drivers of T-cell acute lymphoblastic leukemia (T-ALL). Although a subset of patients with T-ALL display high IL7R messenger RNA levels and cases with IL7R gains have been reported, the impact of IL-7Rα overexpression, rather than mutational activation, during leukemogenesis remains unclear. In this study, overexpressed IL-7Rα in tetracycline-inducible Il7r transgenic and Rosa26 IL7R knockin mice drove potential thymocyte self-renewal, and thymus hyperplasia related to increased proliferation of T-cell precursors, which subsequently infiltrated lymph nodes, spleen, and bone marrow, ultimately leading to fatal leukemia. The tumors mimicked key features of human T-ALL, including heterogeneity in immunophenotype and genetic subtype between cases, frequent hyperactivation of the PI3K/Akt pathway paralleled by downregulation of p27Kip1 and upregulation of Bcl-2, and gene expression signatures evidencing activation of JAK/STAT, PI3K/Akt/mTOR and Notch signaling. Notably, we also found that established tumors may no longer require high levels of IL-7R expression upon secondary transplantation and progressed in the absence of IL-7, but remain sensitive to inhibitors of IL-7R-mediated signaling ruxolitinib (Jak1), AZD1208 (Pim), dactolisib (PI3K/mTOR), palbociclib (Cdk4/6), and venetoclax (Bcl-2). The relevance of these findings for human disease are highlighted by the fact that samples from patients with T-ALL with high wild-type IL7R expression display a transcriptional signature resembling that of IL-7-stimulated pro-T cells and, critically, of IL7R-mutant cases of T-ALL. Overall, our study demonstrates that high expression of IL-7Rα can promote T-cell tumorigenesis, even in the absence of IL-7Rα mutational activation.
Collapse
Affiliation(s)
- Ana Silva
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Afonso R M Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Cachucho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João L Neto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sofie Demeyer
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology
- Katholieke Universiteit (KU) Leuven Center for Human Genetics, Katholieke Universiteit (VIB-KU) Leuven, Leuven, Belgium
| | - Mafalda de Matos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Thea Hogan
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Yunlei Li
- Department of Pathology Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jules Meijerink
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.; and
| | - Jan Cools
- Vlaams Instituut voor Biotechnologie (VIB) Center for Cancer Biology
| | - Ana Rita Grosso
- Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Universidade NOVA de Lisboa, Caparica, Portugal
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, United Kingdom
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
13
|
Liu MY, Klement JD, Langan CJ, van Riggelen J, Liu K. Expression regulation and function of PD-1 and PD-L1 in T lymphoma cells. Cell Immunol 2021; 366:104397. [PMID: 34157461 PMCID: PMC8327398 DOI: 10.1016/j.cellimm.2021.104397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 06/14/2021] [Indexed: 01/01/2023]
Abstract
T lymphoma cells may constitutively express PD-1 and PD-L1. The relative role of PD-1 and PD-L1 in T lymphoma is incompletely understood. We report here that PD-1+ PDL-1+ human T lymphoma cells exhibit constitutive hyperactivation of the TCR signaling and do not respond to PD-L1-mediated suppression in vitro. Knocking out PD-1 or PD-L1 has no effects on T lymphoma cell apoptosis and proliferation in vitro, but significantly increased tumor-bearing mouse survival. Our findings determine that the constitutively active TCR signaling pathway maintain T lymphoma cell growth in vitro and that both PD-1 and PD-L1 promote T lymphoma growth in vivo.
Collapse
Affiliation(s)
- Maria Y Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | - John D Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Candace J Langan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Augusta, GA 30912, USA
| | - Jan van Riggelen
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Augusta, GA 30912, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA; Georgia Cancer Center, Augusta, GA 30912, USA; Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| |
Collapse
|
14
|
Pocock R, Farah N, Richardson SE, Mansour MR. Current and emerging therapeutic approaches for T-cell acute lymphoblastic leukaemia. Br J Haematol 2021; 194:28-43. [PMID: 33942287 DOI: 10.1111/bjh.17310] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
T-cell ALL (T-ALL) is an aggressive malignancy of T-cell progenitors. Although survival outcomes in T-ALL have greatly improved over the past 50 years, relapsed and refractory cases remain extremely challenging to treat and those who cannot tolerate intensive treatment continue to have poor outcomes. Furthermore, T-ALL has proven a more challenging immunotherapeutic target than B-ALL. In this review we explore our expanding knowledge of the basic biology of T-ALL and how this is paving the way for repurposing established treatments and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Rachael Pocock
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Nadine Farah
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Simon E Richardson
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Marc R Mansour
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
15
|
Adenylate kinase 2 expression and addiction in T-ALL. Blood Adv 2021; 5:700-710. [PMID: 33560378 DOI: 10.1182/bloodadvances.2020002700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/16/2020] [Indexed: 01/03/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) represents the malignant expansion of immature T cells blocked in their differentiation. T-ALL is still associated with a poor prognosis, mainly related to occurrence of relapse or refractory disease. A critical medical need therefore exists for new therapies to improve the disease prognosis. Adenylate kinase 2 (AK2) is a mitochondrial kinase involved in adenine nucleotide homeostasis recently reported as essential in normal T-cell development, as defective AK2 signaling pathway results in a severe combined immunodeficiency with a complete absence of T-cell differentiation. In this study, we show that AK2 is constitutively expressed in T-ALL to varying levels, irrespective of the stage of maturation arrest or the underlying oncogenetic features. T-ALL cell lines and patient T-ALL-derived xenografts present addiction to AK2, whereas B-cell precursor ALL cells do not. Indeed, AK2 knockdown leads to early and massive apoptosis of T-ALL cells that could not be rescued by the cytosolic isoform AK1. Mechanistically, AK2 depletion results in mitochondrial dysfunction marked by early mitochondrial depolarization and reactive oxygen species production, together with the depletion of antiapoptotic molecules (BCL-2 and BCL-XL). Finally, T-ALL exposure to a BCL-2 inhibitor (ABT-199 [venetoclax]) significantly enhances the cytotoxic effects of AK2 depletion. We also show that AK2 depletion disrupts the oxidative phosphorylation pathway. Combined with pharmaceutical inhibition of glycolysis, AK2 silencing prevents T-ALL metabolic adaptation, resulting in dramatic apoptosis. Altogether, we pinpoint AK2 as a genuine and promising therapeutic target in T-ALL.
Collapse
|
16
|
Pastorczak A, Domka K, Fidyt K, Poprzeczko M, Firczuk M. Mechanisms of Immune Evasion in Acute Lymphoblastic Leukemia. Cancers (Basel) 2021; 13:1536. [PMID: 33810515 PMCID: PMC8037152 DOI: 10.3390/cancers13071536] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) results from a clonal expansion of abnormal lymphoid progenitors of B cell (BCP-ALL) or T cell (T-ALL) origin that invade bone marrow, peripheral blood, and extramedullary sites. Leukemic cells, apart from their oncogene-driven ability to proliferate and avoid differentiation, also change the phenotype and function of innate and adaptive immune cells, leading to escape from the immune surveillance. In this review, we provide an overview of the genetic heterogeneity and treatment of BCP- and T-ALL. We outline the interactions of leukemic cells in the bone marrow microenvironment, mainly with mesenchymal stem cells and immune cells. We describe the mechanisms by which ALL cells escape from immune recognition and elimination by the immune system. We focus on the alterations in ALL cells, such as overexpression of ligands for various inhibitory receptors, including anti-phagocytic receptors on macrophages, NK cell inhibitory receptors, as well as T cell immune checkpoints. In addition, we describe how developing leukemia shapes the bone marrow microenvironment and alters the function of immune cells. Finally, we emphasize that an immunosuppressive microenvironment can reduce the efficacy of chemo- and immunotherapy and provide examples of preclinical studies showing strategies for improving ALL treatment by targeting these immunosuppressive interactions.
Collapse
Affiliation(s)
- Agata Pastorczak
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, 91-738 Lodz, Poland;
| | - Krzysztof Domka
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Klaudyna Fidyt
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Martyna Poprzeczko
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| | - Malgorzata Firczuk
- Department of Immunology, Medical University of Warsaw, 02-097 Warsaw, Poland; (K.D.); (K.F.); (M.P.)
| |
Collapse
|
17
|
Mshaik R, Simonet J, Georgievski A, Jamal L, Bechoua S, Ballerini P, Bellaye PS, Mlamla Z, Pais de Barros JP, Geissler A, Francin PJ, Girodon F, Garrido C, Quéré R. HSP90 inhibitor NVP-BEP800 affects stability of SRC kinases and growth of T-cell and B-cell acute lymphoblastic leukemias. Blood Cancer J 2021; 11:61. [PMID: 33737511 PMCID: PMC7973815 DOI: 10.1038/s41408-021-00450-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/23/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
T-cell and B-cell acute lymphoblastic leukemias (T-ALL, B-ALL) are aggressive hematological malignancies characterized by an accumulation of immature T- or B-cells. Although patient outcomes have improved, novel targeted therapies are needed to reduce the intensity of chemotherapy and improve the prognosis of high-risk patients. Using cell lines, primary cells and patient-derived xenograft (PDX) models, we demonstrate that ALL cells viability is sensitive to NVP-BEP800, an ATP-competitive inhibitor of Heat shock protein 90 (HSP90). Furthermore, we reveal that lymphocyte-specific SRC family kinases (SFK) are important clients of the HSP90 chaperone in ALL. When PDX mice are treated with NVP-BEP800, we found that there is a decrease in ALL progression. Together, these results demonstrate that the chaperoning of SFK by HSP90 is involved in the growth of ALL. These novel findings provide an alternative approach to target SRC kinases and could be used for the development of new treatment strategies for ALL.
Collapse
Affiliation(s)
- Rony Mshaik
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique de Bourgogne Franche-Comté, Dijon, France
| | - John Simonet
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
| | | | - Layla Jamal
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
| | - Shaliha Bechoua
- Centre de Ressources Biologiques Ferdinand Cabanne, Hôpital Universitaire François Mitterrand, Dijon, France
| | - Paola Ballerini
- Laboratoire d'Hématologie, Assistance Publique Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Pierre-Simon Bellaye
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
- Centre Georges-François Leclerc, Dijon, France
| | - Zandile Mlamla
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
- Plateforme de Lipidomique, Université de Bourgogne Franche-Comté, Dijon, France
| | - Jean-Paul Pais de Barros
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique de Bourgogne Franche-Comté, Dijon, France
- Plateforme de Lipidomique, Université de Bourgogne Franche-Comté, Dijon, France
| | - Audrey Geissler
- Plateforme d'Imagerie Cellulaire, CellImaP, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pierre-Jean Francin
- Laboratoire de Génétique Chromosomique et Moléculaire, Plateau Technique de Biologie, Hôpital Universitaire François Mitterrand, Dijon, France
| | - François Girodon
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
- Service d'Hématologie Biologique, Hôpital Universitaire François Mitterrand, Dijon, France
| | - Carmen Garrido
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique de Bourgogne Franche-Comté, Dijon, France
- Centre Georges-François Leclerc, Dijon, France
| | - Ronan Quéré
- UMR1231, Inserm, Université de Bourgogne Franche-Comté, Dijon, France.
- LipSTIC LabEx, Fondation de Coopération Scientifique de Bourgogne Franche-Comté, Dijon, France.
| |
Collapse
|
18
|
Cieslak A, Charbonnier G, Tesio M, Mathieu EL, Belhocine M, Touzart A, Smith C, Hypolite G, Andrieu GP, Martens JHA, Janssen-Megens E, Gut M, Gut I, Boissel N, Petit A, Puthier D, Macintyre E, Stunnenberg HG, Spicuglia S, Asnafi V. Blueprint of human thymopoiesis reveals molecular mechanisms of stage-specific TCR enhancer activation. J Exp Med 2021; 217:151947. [PMID: 32667968 PMCID: PMC7478722 DOI: 10.1084/jem.20192360] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/03/2020] [Accepted: 05/15/2020] [Indexed: 01/30/2023] Open
Abstract
Cell differentiation is accompanied by epigenetic changes leading to precise lineage definition and cell identity. Here we present a comprehensive resource of epigenomic data of human T cell precursors along with an integrative analysis of other hematopoietic populations. Although T cell commitment is accompanied by large scale epigenetic changes, we observed that the majority of distal regulatory elements are constitutively unmethylated throughout T cell differentiation, irrespective of their activation status. Among these, the TCRA gene enhancer (Eα) is in an open and unmethylated chromatin structure well before activation. Integrative analyses revealed that the HOXA5-9 transcription factors repress the Eα enhancer at early stages of T cell differentiation, while their decommission is required for TCRA locus activation and enforced αβ T lineage differentiation. Remarkably, the HOXA-mediated repression of Eα is paralleled by the ectopic expression of homeodomain-related oncogenes in T cell acute lymphoblastic leukemia. These results highlight an analogous enhancer repression mechanism at play in normal and cancer conditions, but imposing distinct developmental constraints.
Collapse
Affiliation(s)
- Agata Cieslak
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| | - Guillaume Charbonnier
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale, Theories and Approaches of Genomic Complexity, UMR1090, Marseille, France.,Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Melania Tesio
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| | - Eve-Lyne Mathieu
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale, Theories and Approaches of Genomic Complexity, UMR1090, Marseille, France.,Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Mohamed Belhocine
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale, Theories and Approaches of Genomic Complexity, UMR1090, Marseille, France.,Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Aurore Touzart
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France.,Division of Cancer Epigenomics, German Cancer Research Center, Heidelberg, Germany
| | - Charlotte Smith
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| | - Guillaume Hypolite
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| | - Guillaume P Andrieu
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| | - Joost H A Martens
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, Netherlands
| | - Eva Janssen-Megens
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, Netherlands
| | - Marta Gut
- Centro Nacional de Análisis Genómico-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Ivo Gut
- Centro Nacional de Análisis Genómico-Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Nicolas Boissel
- Université Paris Diderot, Institut Universitaire d'Hématologie, EA-3518, Assistance Publique-Hôpitaux de Paris, University Hospital Saint-Louis, Paris, France
| | - Arnaud Petit
- Department of Pediatric Hematology and Oncology, Assistance Publique-Hôpitaux de Paris, Hôpital Armand Trousseau, Paris, France
| | - Denis Puthier
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale, Theories and Approaches of Genomic Complexity, UMR1090, Marseille, France.,Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Elizabeth Macintyre
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculties of Science and Medicine, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, Netherlands
| | - Salvatore Spicuglia
- Aix-Marseille University, Institut National de la Santé et de la Recherche Médicale, Theories and Approaches of Genomic Complexity, UMR1090, Marseille, France.,Equipe Labellisée Ligue Contre le Cancer, Marseille, France
| | - Vahid Asnafi
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de la Santé et de la Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants-Malades, Paris, France
| |
Collapse
|
19
|
Cordo' V, van der Zwet JC, Canté-Barrett K, Pieters R, Meijerink JP. T-cell Acute Lymphoblastic Leukemia: A Roadmap to Targeted Therapies. Blood Cancer Discov 2021; 2:19-31. [PMID: 34661151 PMCID: PMC8447273 DOI: 10.1158/2643-3230.bcd-20-0093] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/08/2020] [Accepted: 10/28/2020] [Indexed: 11/16/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy characterized by aberrant proliferation of immature thymocytes. Despite an overall survival of 80% in the pediatric setting, 20% of patients with T-ALL ultimately die from relapsed or refractory disease. Therefore, there is an urgent need for novel therapies. Molecular genetic analyses and sequencing studies have led to the identification of recurrent T-ALL genetic drivers. This review summarizes the main genetic drivers and targetable lesions of T-ALL and gives a comprehensive overview of the novel treatments for patients with T-ALL that are currently under clinical investigation or that are emerging from preclinical research. SIGNIFICANCE T-ALL is driven by oncogenic transcription factors that act along with secondary acquired mutations. These lesions, together with active signaling pathways, may be targeted by therapeutic agents. Bridging research and clinical practice can accelerate the testing of novel treatments in clinical trials, offering an opportunity for patients with poor outcome.
Collapse
|
20
|
Facts and Challenges in Immunotherapy for T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2020; 21:ijms21207685. [PMID: 33081391 PMCID: PMC7589289 DOI: 10.3390/ijms21207685] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL), a T-cell malignant disease that mainly affects children, is still a medical challenge, especially for refractory patients for whom therapeutic options are scarce. Recent advances in immunotherapy for B-cell malignancies based on increasingly efficacious monoclonal antibodies (mAbs) and chimeric antigen receptors (CARs) have been encouraging for non-responding or relapsing patients suffering from other aggressive cancers like T-ALL. However, secondary life-threatening T-cell immunodeficiency due to shared expression of targeted antigens by healthy and malignant T cells is a main drawback of mAb—or CAR-based immunotherapies for T-ALL and other T-cell malignancies. This review provides a comprehensive update on the different immunotherapeutic strategies that are being currently applied to T-ALL. We highlight recent progress on the identification of new potential targets showing promising preclinical results and discuss current challenges and opportunities for developing novel safe and efficacious immunotherapies for T-ALL.
Collapse
|
21
|
Preclinical efficacy of humanized, non–FcγR-binding anti-CD3 antibodies in T-cell acute lymphoblastic leukemia. Blood 2020; 136:1298-1302. [DOI: 10.1182/blood.2019003801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/04/2020] [Indexed: 01/11/2023] Open
Abstract
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy that accounts for ∼20% of ALL cases. Intensive chemotherapy regimens result in cure rates >85% in children and <50% in adults, warranting a search of novel therapeutic strategies. Although immune-based therapies have tremendously improved the treatment of B-ALL and other B-cell malignancies, they are not yet available for T-ALL. We report here that humanized, non–Fcγ receptor (FcγR)–binding monoclonal antibodies (mAbs) to CD3 have antileukemic properties in xenograft (PDX) models of CD3+ T-ALL, resulting in prolonged host survival. We also report that these antibodies cooperate with chemotherapy to enhance antileukemic effects and host survival. Because these antibodies show only minor, manageable adverse effects in humans, they offer a new therapeutic option for the treatment of T-ALL. Our results also show that the antileukemic properties of anti-CD3 mAbs are largely independent of FcγR-mediated pathways in T-ALL PDXs.
Collapse
|
22
|
|
23
|
Garcia EG, Veloso A, Oliveira ML, Allen JR, Loontiens S, Brunson D, Do D, Yan C, Morris R, Iyer S, Garcia SP, Iftimia N, Van Loocke W, Matthijssens F, McCarthy K, Barata JT, Speleman F, Taghon T, Gutierrez A, Van Vlierberghe P, Haas W, Blackburn JS, Langenau DM. PRL3 enhances T-cell acute lymphoblastic leukemia growth through suppressing T-cell signaling pathways and apoptosis. Leukemia 2020; 35:679-690. [PMID: 32606318 PMCID: PMC8009053 DOI: 10.1038/s41375-020-0937-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 06/10/2020] [Accepted: 06/16/2020] [Indexed: 01/06/2023]
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes and is largely driven by the NOTCH/MYC pathway. Yet, additional oncogenic drivers are required for transformation. Here, we identify protein tyrosine phosphatase type 4 A3 (PRL3) as a collaborating oncogenic driver in T-ALL. PRL3 is expressed in a large fraction of primary human T-ALLs and is commonly co-amplified with MYC. PRL3 also synergized with MYC to initiate early-onset ALL in transgenic zebrafish and was required for human T-ALL growth and maintenance. Mass spectrometry phosphoproteomic analysis and mechanistic studies uncovered that PRL3 suppresses downstream T cell phosphorylation signaling pathways, including those modulated by VAV1, and subsequently suppresses apoptosis in leukemia cells. Taken together, our studies have identified new roles for PRL3 as a collaborating oncogenic driver in human T-ALL and suggest that therapeutic targeting of the PRL3 phosphatase will likely be a useful treatment strategy for T-ALL.
Collapse
Affiliation(s)
- E G Garcia
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - A Veloso
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - M L Oliveira
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - J R Allen
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - S Loontiens
- Cancer Research Institute Ghent, Ghent, Belgium
| | - D Brunson
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - D Do
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - C Yan
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - R Morris
- Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA
| | - S Iyer
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - S P Garcia
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - N Iftimia
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - W Van Loocke
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Biomolecular Medicine and Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - F Matthijssens
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Biomolecular Medicine and Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - K McCarthy
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - J T Barata
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - F Speleman
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Biomolecular Medicine and Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - T Taghon
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - A Gutierrez
- Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, USA
| | - P Van Vlierberghe
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Biomolecular Medicine and Center for Medical Genetics, Ghent University, Ghent, Belgium
| | - W Haas
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA.,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA.,Harvard Stem Cell Institute, Boston, MA, 02114, USA.,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - J S Blackburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - D M Langenau
- Department of Pathology, Massachusetts General Research Institute, Boston, MA, 02114, USA. .,Center of Cancer Research, Massachusetts General Hospital, Charlestown, MA, 02129, USA. .,Harvard Stem Cell Institute, Boston, MA, 02114, USA. .,Center of Regenerative Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
| |
Collapse
|
24
|
Gianni F, Belver L, Ferrando A. The Genetics and Mechanisms of T-Cell Acute Lymphoblastic Leukemia. Cold Spring Harb Perspect Med 2020; 10:a035246. [PMID: 31570389 PMCID: PMC7050584 DOI: 10.1101/cshperspect.a035246] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy derived from early T-cell progenitors. The recognition of clinical, genetic, transcriptional, and biological heterogeneity in this disease has already translated into new prognostic biomarkers, improved leukemia animal models, and emerging targeted therapies. This work reviews our current understanding of the molecular mechanisms of T-ALL.
Collapse
Affiliation(s)
- Francesca Gianni
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Laura Belver
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
| | - Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University Medical Center, New York, New York 10032, USA
- Department of Pathology, Columbia University Medical Center, New York, New York 10032, USA
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10032, USA
| |
Collapse
|
25
|
Dos Santos NR, Ghysdael J, Tran Quang C. The TCR/CD3 complex in leukemogenesis and as a therapeutic target in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2019; 74:100638. [PMID: 31378701 DOI: 10.1016/j.jbior.2019.100638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/27/2019] [Accepted: 07/04/2019] [Indexed: 06/10/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) arises from T cell precursors and is characterized by expression of many lineage-specific proteins. While T-cell antigen receptor (TCR) signaling and its strength are central for thymocyte development, mature T cell homeostasis and immune responses, their roles in T-ALL remain undetermined. Indeed, in contrast to mouse models, in which absence of TCR or major histocompatibility complex binding does not impact on leukemogenesis, other mouse models suggest that basal or weak signaling drives leukemia development. However, recent reports indicate that strong TCR signaling can be detrimental to leukemic cells. Indeed, sustained/high level TCR signaling, stimulated by antigen or CD3 antibody, is strongly anti-leukemic in both murine T-ALL expressing endogenous or transgenic TCR and diagnostic T-ALL cases. As discussed, further work should address the efficacy of T-ALL therapeutic targeting with either TCR/CD3 antibodies or TCR-directed chimeric antigen receptor T cells.
Collapse
Affiliation(s)
- Nuno R Dos Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), 4200-135, Porto, Portugal.
| | - Jacques Ghysdael
- Institut Curie, PSL Research University, CNRS UMR 3348, F-91405, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405, Orsay, France.
| | - Christine Tran Quang
- Institut Curie, PSL Research University, CNRS UMR 3348, F-91405, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405, Orsay, France.
| |
Collapse
|
26
|
Current perspectives in T-ALL. Hemasphere 2019; 3:HemaSphere-2019-0092. [PMID: 35309787 PMCID: PMC8925672 DOI: 10.1097/hs9.0000000000000259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Indexed: 11/26/2022] Open
|
27
|
Abstract
Progress in our understanding of the central genes, pathways, and mechanisms in the pathobiology of T-cell acute lymphoblastic leukemia (T-ALL) has identified key drivers of the disease, opening new opportunities for therapy. Drugs targeting highly prevalent genetic alterations in NOTCH1 and CDKN2A are being explored, and multiple other targets with readily available therapeutic agents, and immunotherapies are being investigated. The molecular basis of T-ALL is reviewed here and potential targets and therapeutic targets discussed.
Collapse
Affiliation(s)
- Adolfo Ferrando
- Institute for Cancer Genetics, Columbia University, 1130 St Nicholas Ave., ICRC 401B, New York, NY, 10032, USA.
| |
Collapse
|
28
|
The depletion of PHF6 decreases the drug sensitivity of T-cell acute lymphoblastic leukemia to prednisolone. Biomed Pharmacother 2018; 109:2210-2217. [PMID: 30551478 DOI: 10.1016/j.biopha.2018.11.083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/18/2018] [Accepted: 11/20/2018] [Indexed: 12/19/2022] Open
Abstract
Mutation of PHF6 has been identified in Börjeson-Forssman-Lehmann syndrome and some types of subsets of childhood leukemia. However, the molecular function and the relationship of PHF6 mutation with glucocorticoid drug resistance during T-ALL treatment remains elusive. Here we report the influence of PHF6 expression on the drug response of T-ALL to prednisolone, and the underlying mechanism of this. Through sanger sequencing and western blotting assays, we identified two T-ALL cell lines with wild-type PHF6 expression, including SIL-ALL and CCRF-CEM, and two T-ALL cell lines without PHF6 expression, including TALL-1 and HPB-ALL, due to the nonsense and frameshift mutations in the coding region of PHF6. MTT assays showed that SIL-ALL and CCRF-CEM were much more sensitive to prednisolone. However, TALL-1 and HPB-ALL were much more resistance to prednisolone. Further knockout of PHF6 led to the resistant of both SIL-ALL and CCRF-CEM cells to prednisolone. On the contrary, the correction of the PHF6 point mutation in HPB-ALL cells with CRISPR-CAS9 method increased the sensibility of both cell lines to prednisolone. Then we found that PHF6 repress p21 expression through direct binding and recruiting RBPP4 to its promoter region. Finally, the co-treatment of p21 inhibitor increased the sensitivity of TALL-1 and HPB-ALL cells to prednisolone. Collectively, our findings not only enrich our understanding of the relationship between PHF6 mutation and drug resistance but also indicate a new therapeutic potential for those T-ALL patients containing the PHF6 mutation.
Collapse
|
29
|
Bassan R, Bourquin JP, DeAngelo DJ, Chiaretti S. New Approaches to the Management of Adult Acute Lymphoblastic Leukemia. J Clin Oncol 2018; 36:JCO2017773648. [PMID: 30240326 DOI: 10.1200/jco.2017.77.3648] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Traditional treatment regimens for adult acute lymphoblastic leukemia, including allogeneic hematopoietic cell transplantation, result in an overall survival of approximately 40%, a figure hardly comparable with the extraordinary 80% to 90% cure rate currently reported in children. When translated to the adult setting, modern pediatric-type regimens improve the survival to approximately 60% in young adults. The addition of tyrosine kinase inhibitors for patients with Philadelphia chromosome-positive disease and the measurement of minimal residual disease to guide risk stratification and postremission approaches has led to additional improvements in outcomes. Relapsed disease and treatment toxicity-sparing no patient but representing a major concern especially in the elderly-are the most critical current issues awaiting further therapeutic advancement. Recently, there has been considerable progress in understanding the disease biology, specifically the Philadelphia-like signature, as well as other high-risk subgroups. In addition, there are several new agents that will undoubtedly contribute to additional improvement in the current outcomes. The most promising agents are monoclonal antibodies, immunomodulators, and chimeric antigen receptor T cells, and, to a lesser extent, several new drugs targeting key molecular pathways involved in leukemic cell growth and proliferation. This review examines the evidence supporting the increasing role of the new therapeutic tools and treatment options in different disease subgroups, including frontline and relapsed or refractory disease. It is now possible to define the best individual approach on the basis of the emerging concepts of precision medicine.
Collapse
Affiliation(s)
- Renato Bassan
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| | - Jean-Pierre Bourquin
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| | - Daniel J DeAngelo
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| | - Sabina Chiaretti
- Renato Bassan, Ospedale dell'Angelo, Mestre-Venezia; Sabina Chiaretti, "Sapienza" University, Rome, Italy; Jean-Pierre Bourquin, University Children's Hospital, Zurich, Switzerland; and Daniel J. DeAngelo, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
30
|
Gon S, Loosveld M, Crouzet T, Potier D, Bonnet M, Morin SO, Michel G, Vey N, Nunès JA, Malissen B, Roncagalli R, Nadel B, Payet-Bornet D. Fit αβ T-cell receptor suppresses leukemogenesis of Pten-deficient thymocytes. Haematologica 2018; 103:999-1007. [PMID: 29567770 PMCID: PMC6058769 DOI: 10.3324/haematol.2018.188359] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/15/2018] [Indexed: 12/25/2022] Open
Abstract
Signaling through the αβT cell receptor (TCR) is a crucial determinant of T-cell fate and can induce two opposite outcomes during thymocyte development: cell death or survival and differentiation. To date, the role played by T-cell receptor in the oncogenic transformation of developing T cells remains unclear. Here we show that human primary T-cell acute lymphoblastic leukemias expressing an αβT cell receptor are frequently deficient for phosphatase and tensin homolog protein (PTEN), and fail to respond strongly to T-cell receptor activation. Using Pten-deficient T-cell acute lymphoblastic leukemia mouse models, we confirm that T-cell receptor signaling is involved in leukemogenesis. We show that abrogation of T-cell receptor expression accelerated tumor onset, while enforced expression of a fit transgenic T-cell receptor led to the development of T-cell receptor-negative lymphoma and delayed tumorigenesis. We further demonstrate that pre-tumoral Pten-deficient thymocytes harboring fit T-cell receptors undergo early clonal deletion, thus preventing their malignant transformation, while cells with unfit T-cell receptors that should normally be deleted during positive selection, pass selection and develop T-cell acute lymphoblastic leukemias. Altogether, our data show that fit T-cell receptor signaling suppresses tumor development mediated by Pten loss-of-function and point towards a role of Pten in positive selection.
Collapse
Affiliation(s)
- Stéphanie Gon
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Marie Loosveld
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France.,APHM, Hôpital La Timone, Laboratoire d'Hématologie, CRCM, Marseille, France
| | - Thomas Crouzet
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Delphine Potier
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Mélanie Bonnet
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Stéphanie O Morin
- Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Gérard Michel
- APHM, Hôpital La Timone, Service d'Hématologie et d'Oncologie Pédiatrique, Marseille, France
| | - Norbert Vey
- Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.,Institut Paoli-Calmettes, Hematology Department, Marseille, France
| | - Jacques A Nunès
- Aix-Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | | | - Bertrand Nadel
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|
31
|
Abstract
Thymic negative selection is a process that aims to eliminate autoreactive T cells by inducing the apoptosis of thymocytes expressing a T-cell receptor (TCR) with high affinity for self-MHC. In this issue, Trinquand and colleagues demonstrate that TCR engagement or anti-CD3 stimulation of TCR-expressing T acute lymphoblastic leukemia cells results in their apoptosis. This cell death is reminiscent of thymic negative selection and has the potential for therapeutic exploitation. Cancer Discov; 6(9); 946-8. ©2016 AACR.See related article by Trinquand et al., p. 972.
Collapse
Affiliation(s)
- François Lemonnier
- Campbell Family Institute for Breast Cancer Research at the Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research at the Princess Margaret Cancer Centre, University Health Network, Toronto, Canada. Department of Medical Biophysics, University of Toronto, University Health Network, Toronto, Canada.
| |
Collapse
|
32
|
Oliveira ML, Akkapeddi P, Alcobia I, Almeida AR, Cardoso BA, Fragoso R, Serafim TL, Barata JT. From the outside, from within: Biological and therapeutic relevance of signal transduction in T-cell acute lymphoblastic leukemia. Cell Signal 2017. [PMID: 28645565 DOI: 10.1016/j.cellsig.2017.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer that arises from clonal expansion of transformed T-cell precursors. In this review we summarize the current knowledge on the external stimuli and cell-intrinsic lesions that drive aberrant activation of pivotal, pro-tumoral intracellular signaling pathways in T-cell precursors, driving transformation, leukemia expansion, spread or resistance to therapy. In addition to their pathophysiological relevance, receptors and kinases involved in signal transduction are often attractive candidates for targeted drug development. As such, we discuss also the potential of T-ALL signaling players as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mariana L Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Padma Akkapeddi
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Isabel Alcobia
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Afonso R Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno A Cardoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Rita Fragoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Teresa L Serafim
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
33
|
Girardi T, Vicente C, Cools J, De Keersmaecker K. The genetics and molecular biology of T-ALL. Blood 2017; 129:1113-1123. [PMID: 28115373 PMCID: PMC5363819 DOI: 10.1182/blood-2016-10-706465] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/10/2016] [Indexed: 12/13/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy caused by the accumulation of genomic lesions that affect the development of T cells. For many years, it has been established that deregulated expression of transcription factors, impairment of the CDKN2A/2B cell-cycle regulators, and hyperactive NOTCH1 signaling play prominent roles in the pathogenesis of this leukemia. In the past decade, systematic screening of T-ALL genomes by high-resolution copy-number arrays and next-generation sequencing technologies has revealed that T-cell progenitors accumulate additional mutations affecting JAK/STAT signaling, protein translation, and epigenetic control, providing novel attractive targets for therapy. In this review, we provide an update on our knowledge of T-ALL pathogenesis, the opportunities for the introduction of targeted therapy, and the challenges that are still ahead.
Collapse
Affiliation(s)
- Tiziana Girardi
- Department of Oncology, KU Leuven, Leuven, Belgium
- Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Carmen Vicente
- Leuven Cancer Institute (LKI), Leuven, Belgium
- VIB Center for the Biology of Disease, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Jan Cools
- Leuven Cancer Institute (LKI), Leuven, Belgium
- VIB Center for the Biology of Disease, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Kim De Keersmaecker
- Department of Oncology, KU Leuven, Leuven, Belgium
- Leuven Cancer Institute (LKI), Leuven, Belgium
| |
Collapse
|
34
|
Quang CT, Zaniboni B, Ghysdael J. A TCR-switchable cell death pathway in T-ALL. Oncoscience 2017; 4:17-18. [PMID: 28540327 PMCID: PMC5441467 DOI: 10.18632/oncoscience.342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 03/07/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- Christine Tran Quang
- Institut Curie, PSL Research University, CNRS UMR 3348, F-91405, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405 Orsay, France
| | - Benedetta Zaniboni
- Institut Curie, PSL Research University, CNRS UMR 3348, F-91405, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405 Orsay, France
| | - Jacques Ghysdael
- Institut Curie, PSL Research University, CNRS UMR 3348, F-91405, Orsay, France; Université Paris Sud, Université Paris-Saclay, CNRS UMR 3348, F-91405 Orsay, France
| |
Collapse
|
35
|
Karrman K, Johansson B. Pediatric T-cell acute lymphoblastic leukemia. Genes Chromosomes Cancer 2016; 56:89-116. [PMID: 27636224 DOI: 10.1002/gcc.22416] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/06/2016] [Indexed: 12/29/2022] Open
Abstract
The most common pediatric malignancy is acute lymphoblastic leukemia (ALL), of which T-cell ALL (T-ALL) comprises 10-15% of cases. T-ALL arises in the thymus from an immature thymocyte as a consequence of a stepwise accumulation of genetic and epigenetic aberrations. Crucial biological processes, such as differentiation, self-renewal capacity, proliferation, and apoptosis, are targeted and deranged by several types of neoplasia-associated genetic alteration, for example, translocations, deletions, and mutations of genes that code for proteins involved in signaling transduction, epigenetic regulation, and transcription. Epigenetically, T-ALL is characterized by gene expression changes caused by hypermethylation of tumor suppressor genes, histone modifications, and miRNA and lncRNA abnormalities. Although some genetic and gene expression patterns have been associated with certain clinical features, such as immunophenotypic subtype and outcome, none has of yet generally been implemented in clinical routine for treatment decisions. The recent advent of massive parallel sequencing technologies has dramatically increased our knowledge of the genetic blueprint of T-ALL, revealing numerous fusion genes as well as novel gene mutations. The challenges now are to integrate all genetic and epigenetic data into a coherent understanding of the pathogenesis of T-ALL and to translate the wealth of information gained in the last few years into clinical use in the form of improved risk stratification and targeted therapies. Here, we provide an overview of pediatric T-ALL with an emphasis on the acquired genetic alterations that result in this disease. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kristina Karrman
- Department of Clinical Genetics, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Bertil Johansson
- Department of Clinical Genetics, Office for Medical Services, Division of Laboratory Medicine, Lund, Sweden.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|