1
|
Bakhtiari M, Jordan SC, Mumme HL, Sharma R, Shanmugam M, Bhasin SS, Bhasin M. ARMH1 is a novel marker associated with poor pediatric AML outcomes that affect the fatty acid synthesis and cell cycle pathways. Front Oncol 2024; 14:1445173. [PMID: 39703843 PMCID: PMC11655347 DOI: 10.3389/fonc.2024.1445173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Despite remarkable progress in Pediatric Acute Myeloid Leukemia (pAML) treatments, the relapsed disease remains difficult to treat, making it pertinent to identify novel biomarkers of prognostic/therapeutic significance. Material and methods Bone marrow samples from 21 pAML patients were analyzed using single cell RNA sequencing, functional assays with ARMH1 knockdown and overexpression were performed in leukemia cell lines to evaluate impact on proliferation and migration, and chemotherapy sensitivity. Mitochondrial function was assessed via Seahorse assay, ARMH1 interacting proteins were studied using co-immunoprecipitation. Bulk RNA-seq was performed on ARMH1knockdown and over expressing cell lines to evaluate the pathways and networks impacted by ARMH1. Results Our data shows that ARMH1, a novel cancer-associated gene, is highly expressed in the malignant blast cells of multiple pediatric hematologic malignancies, including AML, T/B-ALL, and T/B-MPAL. Notably, ARMH1 expression is significantly elevated in blast cells of patients who relapsed or have a high-risk cytogenetic profile (MLL) compared to standard-risk (RUNX1, inv (16)). ARMH1 expression is also significantly correlated with the pediatric leukemia stem cell score of 6 genes (LSC6) associated with poor outcomes. Perturbation of ARMH1 (knockdown and overexpression) in leukemia cell lines significantly impacted cell proliferation and migration. The RNA-sequencing analysis on multiple ARMH1 knockdown and overexpressing cell lines established an association with mitochondrial fatty acid synthesis and cell cycle pathways.The investigation of the mitochondrial matrix shows that pharmacological inhibition of a key enzyme in fatty acid synthesis regulation, CPT1A, resulted in ARMH1 downregulation. ARMH1 knockdown also led to a significant reduction in CPT1A and ATP production as well as Oxygen Consumption Rate. Our data indicates that downregulating ARMH1 impacts cell proliferation by reducing key cell cycle regulators such as CDCA7 and EZH2. Further, we also established that ARMH1 is a key physical interactant of EZH2, associated with multiple cancers. Conclusion Our findings underscore further evaluation of ARMH1 as a potential candidate for targeted therapies and stratification of aggressive pAML to improve outcomes.
Collapse
Affiliation(s)
- Mojtaba Bakhtiari
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
| | - Sean C. Jordan
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Hope L. Mumme
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
| | - Richa Sharma
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Swati S. Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| | - Manoj Bhasin
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA, United States
- Department of Biomedical Informatics, Emory University, Atlanta, GA, United States
- Department of Pediatrics, Emory University, Atlanta, GA, United States
| |
Collapse
|
2
|
Fan Y, Sun L, He J, Chen Y, Ma H, Ding H. Siglec15 in blood system diseases: from bench to bedside. Front Immunol 2024; 15:1490505. [PMID: 39697338 PMCID: PMC11652361 DOI: 10.3389/fimmu.2024.1490505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting the PD-1/PD-L1 pathway using immunomodulators has demonstrated promising outcomes in clinics. Immunomodulators can effectively target immune checkpoints with a strong preference for the tumor microenvironment (TME). Besides, immunomodulators specifically target the recently discovered inhibitory immune checkpoint, sialic acid-binding immunoglobulin-like lectin (Siglec-15). Distinctive in its molecular composition, Siglec-15 has a unique molecular composition and been shown to be highly prevalent in numerous solid tumor tissues and tumor-associated macrophages (TAMs) in human subjects. Notably, Siglec-15 is up-regulated across various cancer types. As a result, Siglec-15 has attracted significant attention due to its exclusive nature concerning PD-L1 expression, suggesting its role in immune evasion in patients lacking PD-L1. Siglec-15 predominantly appears in certain populations and can promote tumor development by repressing T lymphocyte activation and proliferation, thereby facilitating tumor cell immune escape. Furthermore, Siglec-15 is implicated in osteoclast differentiation and bone remodeling, indicating that it is a promising target for next-generation cancer immunotherapies. Additionally, Siglec-15 can modulate immune responses to microbial infections. The current treatment strategies for hematological conditions predominantly include conventional intensive chemotherapy and transplantation methods. However, emerging immunotherapeutic approaches are increasingly recognized for their overall effectiveness, indicating that specific molecular targets should be identified. The expression of Siglec-15 within tumor cells may indicate a novel pathway for treating hematological malignancies. In this study, the biological attributes, expression patterns, and pathogenic mechanisms of Siglec-15 across various diseases were reviewed. The role of Siglec-15 in the pathogenesis and laboratory diagnosis of hematological disorders was also evaluated.
Collapse
Affiliation(s)
- Yujia Fan
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Liangliang Sun
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Juan He
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yuetong Chen
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Hongli Ma
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Haitao Ding
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Danylesko I, Shem-Tov N, Yerushalmi R, Jacoby E, Toren A, Shouval R, Itzhaki O, Avigdor A, Shimoni A, Nagler A. Point of care CD19 chimeric antigen receptor (CAR) T-cells for relapsed/refractory acute myeloid leukemia (AML) with aberrant CD19 antigen expression. Curr Res Transl Med 2024; 72:103471. [PMID: 39305562 DOI: 10.1016/j.retram.2024.103471] [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: 05/09/2024] [Revised: 08/10/2024] [Accepted: 09/14/2024] [Indexed: 12/07/2024]
Abstract
Relapsed/refractory (r/r) acute myeloid leukemia (AML) is associated with poor prognosis. CD19 is a B-cell marker, is aberrantly expressed in AML, mostly with t(8; 21)(q22; q22.1). Here we report the results of a phase 2 study giving point of care produced CD19 CAR T- cells for r/r AML with aberrant expression of CD19 (NCT04257175). Lymphodepletion included fludarabine and cyclophosphamide The response was evaluated by bone marrow (BM) aspiration on day 28. Six patients (5 adults and 1 child) were included. Median number of previous chemotherapy lines was 4 (range, 3-8) and four patients received CAR T-cells 8-18 months post allogeneic hematopoietic stem cell transplantation (allo-HSCT). Cytokine release syndrome (CRS) of any grade occurred in all patients, and 1 patient had grade 3 CRS. Immune effector cell-associated neurotoxicity syndrome (ICANS) occurred in 2 patients at low grades. Tocilizumab was administered to 2 patients and corticosteroids to 3 patients. Four patients achieved a complete remission (CR), while 2/6 progressed (PD). Three patients (2 with CR and 1 with PD) underwent allo-HSCT (it was the second transplant in 2) 2-5 months post CAR T-cells infusion. The median duration of response in patients achieving CR was 8.5 (range; 3-14) months. However, all patients eventually died within 5 (1-18) months. In conclusion, CD19 CAR T- cell treatment for AML is feasible and safe. However, the response is short and should be followed by allo-HSCT. Hopefully, future long term results will be improved by combining the CAR T- cell therapy with the emerging novel effective anti-leukemic compounds.
Collapse
Affiliation(s)
- Ivetta Danylesko
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Noga Shem-Tov
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Yerushalmi
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elad Jacoby
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Amos Toren
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Roni Shouval
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Department of Medicine, Weill Cornell Medical College, New York, New York, USA; Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Orit Itzhaki
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Ella Lemelbaum Institute of Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Abraham Avigdor
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avichai Shimoni
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Arnon Nagler
- Division of Hematology and Bone Marrow Transplantation, Sheba Medical Center, Ramat Gan, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Casirati G. To target, to escape, perchance to cure: Borrowing a page from cancer's playbook, scientists learn to evade their own therapies. Science 2024; 386:739. [PMID: 39541456 DOI: 10.1126/science.adt9029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Borrowing a page from cancer's playbook, scientists learn to evade their own therapies.
Collapse
Affiliation(s)
- Gabriele Casirati
- Dana-Farber Cancer Institute, Boston, MA, USA
- Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
5
|
Ji RJ, Cao GH, Zhao WQ, Wang MY, Gao P, Zhang YZ, Wang XB, Qiu HY, Chen DD, Tong XH, Duan M, Yin H, Zhang Y. Epitope prime editing shields hematopoietic cells from CD123 immunotherapy for acute myeloid leukemia. Cell Stem Cell 2024; 31:1650-1666.e8. [PMID: 39353428 DOI: 10.1016/j.stem.2024.09.003] [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: 11/28/2023] [Revised: 07/28/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Acute myeloid leukemia (AML) is a malignant cancer characterized by abnormal differentiation of hematopoietic stem and progenitor cells (HSPCs). While chimeric antigen receptor T (CAR-T) cell immunotherapies target AML cells, they often induce severe on-target/off-tumor toxicity by attacking normal cells expressing the same antigen. Here, we used base editors (BEs) and a prime editor (PE) to modify the epitope of CD123 on HSPCs, protecting healthy cells from CAR-T-induced cytotoxicity while maintaining their normal function. Although BE effectively edits epitopes, complex bystander products are a concern. To enhance precision, we optimized prime editing, increasing the editing efficiency from 5.9% to 78.9% in HSPCs. Epitope-modified cells were resistant to CAR-T lysis while retaining normal differentiation and function. Furthermore, BE- or PE-edited HSPCs infused into humanized mice endowed myeloid lineages with selective resistance to CAR-T immunotherapy, demonstrating a proof-of-concept strategy for treating relapsed AML.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Humans
- Interleukin-3 Receptor alpha Subunit/metabolism
- Animals
- Hematopoietic Stem Cells/immunology
- Hematopoietic Stem Cells/metabolism
- Mice
- Epitopes/immunology
- Immunotherapy/methods
- Gene Editing
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Rui-Jin Ji
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Guo-Hua Cao
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Wei-Qiang Zhao
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Mu-Yao Wang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Pan Gao
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yi-Zhou Zhang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xue-Bin Wang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hou-Yuan Qiu
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Di-Di Chen
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xiao-Han Tong
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Min Duan
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hao Yin
- Departments of Clinical Laboratory and Department of Urology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; TaiKang Centre for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China; State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China
| | - Ying Zhang
- Department of Rheumatology and Immunology, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; TaiKang Centre for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China; State Key Laboratory of Virology, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
6
|
Ravandi F, Subklewe M, Walter RB, Vachhani P, Ossenkoppele G, Buecklein V, Döhner H, Jongen-Lavrencic M, Baldus CD, Fransecky L, Pardee TS, Kantarjian H, Yen PK, Mukundan L, Panwar B, Yago MR, Agarwal S, Khaldoyanidi SK, Stein A. Safety and tolerability of AMG 330 in adults with relapsed/refractory AML: a phase 1a dose-escalation study. Leuk Lymphoma 2024; 65:1281-1291. [PMID: 38712673 DOI: 10.1080/10428194.2024.2346755] [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: 11/21/2023] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/08/2024]
Abstract
AMG 330, a bispecific T-cell engager (BiTE®) that binds CD33 and CD3 on T cells facilitates T-cell-mediated cytotoxicity against CD33+ cells. This first-in-human, open-label, dose-escalation study evaluated the safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of AMG 330 in adults with relapsed/refractory acute myeloid leukemia (R/R AML). Amongst 77 patients treated with AMG 330 (0.5 µg/day-1.6 mg/day) on 14-day or 28-day cycles, maximum tolerated dose was not reached; median duration of treatment was 29 days. The most frequent treatment-related adverse events were cytokine release syndrome (CRS; 78%) and rash (30%); 10% of patients experienced grade 3/4 CRS. CRS was mitigated with stepwise dosing of AMG 330, prophylactic dexamethasone, and early treatment with tocilizumab. Among 60 evaluable patients, eight achieved complete remission or morphologic leukemia-free state; of the 52 non-responders, 37% had ≥50% reduction in AML bone marrow blasts. AMG 330 is a promising CD33-targeted therapeutic strategy for R/R AML.
Collapse
MESH Headings
- Humans
- Male
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/diagnosis
- Female
- Middle Aged
- Adult
- Aged
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/adverse effects
- Antibodies, Bispecific/therapeutic use
- Treatment Outcome
- Young Adult
- Maximum Tolerated Dose
- Drug Resistance, Neoplasm/drug effects
- Sialic Acid Binding Ig-like Lectin 3/metabolism
- Recurrence
- Aged, 80 and over
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Dose-Response Relationship, Drug
- Cytokine Release Syndrome/etiology
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Research, Gene Center, LMU Munich, Munich, Germany
| | - Roland B Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Pankit Vachhani
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Veit Buecklein
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Research, Gene Center, LMU Munich, Munich, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Mojca Jongen-Lavrencic
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Claudia D Baldus
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lars Fransecky
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Timothy S Pardee
- Department of Internal Medicine, Section on Hematology and Oncology, Atrium Health Wake Forest Baptist, Winston-Salem, NC, USA
| | - Hagop Kantarjian
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | - Anthony Stein
- Gehr Family Center for Leukemia Research, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
7
|
Sharifi MJ, Xu L, Nasiri N, Ashja‐Arvan M, Soleimanzadeh H, Ganjalikhani‐Hakemi M. Immune-dysregulation harnessing in myeloid neoplasms. Cancer Med 2024; 13:e70152. [PMID: 39254117 PMCID: PMC11386321 DOI: 10.1002/cam4.70152] [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: 05/05/2024] [Revised: 08/02/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Myeloid malignancies arise in bone marrow microenvironments and shape these microenvironments in favor of malignant development. Immune suppression is one of the most important stages in myeloid leukemia progression. Leukemic clone expansion and immune dysregulation occur simultaneously in bone marrow microenvironments. Complex interactions emerge between normal immune system elements and leukemic clones in the bone marrow. In recent years, researchers have identified several of these pathological interactions. For instance, recent works shows that the secretion of inflammatory cytokines such as tumor necrosis factor-α (TNF-α), from bone marrow stromal cells contributes to immune dysregulation and the selective proliferation of JAK2V617F+ clones in myeloproliferative neoplasms. Moreover, inflammasome activation and sterile inflammation result in inflamed microenvironments and the development of myelodysplastic syndromes. Additional immune dysregulations, such as exhaustion of T and NK cells, an increase in regulatory T cells, and impairments in antigen presentation are common findings in myeloid malignancies. In this review, we discuss the role of altered bone marrow microenvironments in the induction of immune dysregulations that accompany myeloid malignancies. We also consider both current and novel therapeutic strategies to restore normal immune system function in the context of myeloid malignancies.
Collapse
Affiliation(s)
- Mohammad Jafar Sharifi
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical SciencesShiraz University of Medical SciencesShirazIran
| | - Ling Xu
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan UniversityGuangzhouChina
| | - Nahid Nasiri
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical SciencesShiraz University of Medical SciencesShirazIran
| | - Mehnoosh Ashja‐Arvan
- Regenerative and Restorative Medicine Research Center (REMER)Research Institute of Health sciences and Technology (SABITA), Istanbul Medipol UniversityIstanbulTurkey
| | - Hadis Soleimanzadeh
- Division of Laboratory Hematology and Blood Banking, Department of Medical Laboratory Sciences, School of Paramedical SciencesShiraz University of Medical SciencesShirazIran
| | - Mazdak Ganjalikhani‐Hakemi
- Regenerative and Restorative Medicine Research Center (REMER)Research Institute of Health sciences and Technology (SABITA), Istanbul Medipol UniversityIstanbulTurkey
- Department of Immunology, Faculty of MedicineIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
8
|
Hayatigolkhatmi K, Valzelli R, El Menna O, Minucci S. Epigenetic alterations in AML: Deregulated functions leading to new therapeutic options. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 387:27-75. [PMID: 39179348 DOI: 10.1016/bs.ircmb.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
Acute myeloid leukemia (AML) results in disruption of the hematopoietic differentiation process. Crucial progress has been made, and new therapeutic strategies for AML have been developed. Induction chemotherapy, however, remains the main option for the majority of AML patients. Epigenetic dysregulation plays a central role in AML pathogenesis, supporting leukemogenesis and maintenance of leukemic stem cells. Here, we provide an overview of the intricate interplay of altered epigenetic mechanisms, including DNA methylation, histone modifications, and chromatin remodeling, in AML development. We explore the role of epigenetic regulators, such as DNMTs, HMTs, KDMs, and HDACs, in mediating gene expression patterns pushing towards leukemic cell transformation. Additionally, we discuss the impact of cytogenetic lesions on epigenomic remodeling and the potential of targeting epigenetic vulnerabilities as a therapeutic strategy. Understanding the epigenetic landscape of AML offers insights into novel therapeutic avenues, including epigenetic modifiers and particularly their use in combination therapies, to improve treatment outcomes and overcome drug resistance.
Collapse
Affiliation(s)
- Kourosh Hayatigolkhatmi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Riccardo Valzelli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Oualid El Menna
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy; Department of Hemato-Oncology, Università Statale di Milano, Milan, Italy.
| |
Collapse
|
9
|
Restelli C, Ruella M, Paruzzo L, Tarella C, Pelicci PG, Colombo E. Recent Advances in Immune-Based Therapies for Acute Myeloid Leukemia. Blood Cancer Discov 2024; 5:234-248. [PMID: 38904305 PMCID: PMC11215380 DOI: 10.1158/2643-3230.bcd-23-0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/16/2024] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Despite advancements, acute myeloid leukemia (AML) remains unconquered by current therapies. Evidence of immune evasion during AML progression, such as HLA loss and T-cell exhaustion, suggests that antileukemic immune responses contribute to disease control and could be harnessed by immunotherapy. In this review, we discuss a spectrum of AML immunotherapy targets, encompassing cancer cell-intrinsic and surface antigens as well as targeting in the leukemic milieu, and how they can be tailored for personalized approaches. These targets are overviewed across major immunotherapy modalities applied to AML: immune checkpoint inhibitors, antibody-drug conjugates, therapeutic vaccines, bispecific/trispecific antibodies, and chimeric antigen receptor (CAR)-T and CAR-NK cells. Significance: Immune therapies in AML treatment show evolving promise. Ongoing research aims to customize approaches for varied patient profiles and clinical scenarios. This review covers immune surveillance mechanisms, therapy options like checkpoint inhibitors, antibodies, CAR-T/NK cells, and vaccines, as well as resistance mechanisms and microenvironment considerations.
Collapse
Affiliation(s)
- Cecilia Restelli
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy.
| | - Marco Ruella
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA.
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA.
| | - Luca Paruzzo
- Center for Cellular Immunotherapies and Cellular Therapy and Transplant, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA.
- Division of Hematology-Oncology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA.
| | - Corrado Tarella
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy.
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy.
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy.
| | - Emanuela Colombo
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan, Italy.
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
10
|
Perzolli A, Koedijk JB, Zwaan CM, Heidenreich O. Targeting the innate immune system in pediatric and adult AML. Leukemia 2024; 38:1191-1201. [PMID: 38459166 PMCID: PMC11147779 DOI: 10.1038/s41375-024-02217-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
While the introduction of T cell-based immunotherapies has improved outcomes in many cancer types, the development of immunotherapies for both adult and pediatric AML has been relatively slow and limited. In addition to the need to identify suitable target antigens, a better understanding of the immunosuppressive tumor microenvironment is necessary for the design of novel immunotherapy approaches. To date, most immune characterization studies in AML have focused on T cells, while innate immune lineages such as monocytes, granulocytes and natural killer (NK) cells, received less attention. In solid cancers, studies have shown that innate immune cells, such as macrophages, myeloid-derived suppressor cells and neutrophils are highly plastic and may differentiate into immunosuppressive cells depending on signals received in their microenvironment, while NK cells appear to be functionally impaired. Hence, an in-depth characterization of the innate immune compartment in the TME is urgently needed to guide the development of immunotherapeutic interventions for AML. In this review, we summarize the current knowledge on the innate immune compartment in AML, and we discuss how targeting its components may enhance T cell-based- and other immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alicia Perzolli
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Joost B Koedijk
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands.
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
11
|
Gao C, Li X, Xu Y, Zhang T, Zhu H, Yao D. Recent advances in CAR-T cell therapy for acute myeloid leukaemia. J Cell Mol Med 2024; 28:e18369. [PMID: 38712978 PMCID: PMC11075639 DOI: 10.1111/jcmm.18369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/18/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a fatal and refractory haematologic cancer that primarily affects adults. It interferes with bone marrow cell proliferation. Patients have a 5 years survival rate of less than 30% despite the availability of several treatments, including chemotherapy, allogeneic haematopoietic stem cell transplantation (Allo-HSCT), and receptor antagonist drugs. Allo-HSCT is the mainstay of acute myeloid leukaemia treatment. Although it does work, there are severe side effects, such as graft-versus-host disease (GVHD). In recent years, chimeric antigen receptor (CAR)-T cell therapies have made significant progress in the treatment of cancer. These engineered T cells can locate and recognize tumour cells in vivo and release a large number of effectors through immune action to effectively kill tumour cells. CAR-T cells are among the most effective cancer treatments because of this property. CAR-T cells have demonstrated positive therapeutic results in the treatment of acute myeloid leukaemia, according to numerous clinical investigations. This review highlights recent progress in new targets for AML immunotherapy, and the limitations, and difficulties of CAR-T therapy for AML.
Collapse
Affiliation(s)
- Chi Gao
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Xin Li
- College of BiotechnologyTianjin University of Science and TechnologyTianjinChina
| | - Yao Xu
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Tongcun Zhang
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
- Institute of Biology and MedicineWuhan University of Science and TechnologyWuhanChina
| | - Haichuan Zhu
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Di Yao
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| |
Collapse
|
12
|
Zhao Y, Zhang X, Zhang M, Guo R, Zhang Y, Pu Y, Zhu H, Liu P, Zhang Y, He X, Lyu C, Lyu H, Xiao X, Zhao M. Modified EASIX scores predict severe CRS/ICANS in patients with acute myeloid leukemia following CLL1 CAR-T cell therapy. Ann Hematol 2024; 103:969-980. [PMID: 38214708 DOI: 10.1007/s00277-024-05617-y] [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: 10/25/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy targeting CLL1 has been considered a potent weapon for patients with acute myeloid leukemia (AML). This study aims to evaluate the efficacy and toxicity of CLL1 CAR-T cell therapy in a larger cohort, with particular attention to cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Among the 32 patients assessed for efficacy, complete remission occurred in 71.88% (23/32) of cases and undetectable minimal residual disease in 14 patients. The CRS developed in all patients, with 8 individuals experiencing ICANS. Severe CRS and ICANS were observed in 11 and 2 patients, respectively. Furthermore, the Endothelial Activation and Stress Index (EASIX) and its derivatives measured before and after CLL1 CAR-T cell infusion were employed for predicting the severe complications. Significant differences were observed in EASIX scores on the day before lymphodepletion (Day BL, P = 0.023), -1 (P < 0.001), +1 (P < 0.001), and +3(P = 0.014); sEASIX scores on Day BL (P = 0.007), -1 (P < 0.001), +1 (P < 0.001), and +3 (P < 0.001); and mEASIX score on Day -1 (P = 0.004) between patients with mild and severe CRS/ICANS. Additionally, there was a significant difference in mEASIX scores between responders and non-responders on Day BL (P = 0.004) and Day -1 (P = 0.044). Our findings indicate that pre- and post-infusion assessments of EASIX/mEASIX/sEASIX scores serve as reliable prognostic indicators for severe CRS/ICANS and treatment response following CLL1 CAR-T cell therapy, which can assist physicians in implementing preemptive treatment strategies for potential severe complications and screening patients who are suitable candidates for CLL1 CAR-T cell therapy. EASIX/mEASIX/sEASIX scores serve as reliable prognostic indicators for severe CRS/ICANS following CLL1 CAR-T cell therapy. The preinfusion mEASIX scores of CLL1 CAR-T cells can effectively predict treatment response.
Collapse
Affiliation(s)
- Yifan Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Xiaomei Zhang
- Nankai University School of Medicine, Tianjin, 300380, China
| | - Meng Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Ruiting Guo
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Yi Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, 300380, China
| | - Yedi Pu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Haibo Zhu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Pengjiang Liu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Yu Zhang
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Xiaoyuan He
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Cuicui Lyu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Hairong Lyu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China
| | - Xia Xiao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China.
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300380, China.
| |
Collapse
|
13
|
Clarke ML, Gabrielsen OS, Frampton J. MYB as a Critical Transcription Factor and Potential Therapeutic Target in AML. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:341-358. [PMID: 39017851 DOI: 10.1007/978-3-031-62731-6_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Myb was identified over four decades ago as the transforming component of acute leukemia viruses in chickens. Since then it has become increasingly apparent that dysregulated MYB activity characterizes many blood cancers, including acute myeloid leukemia, and that it represents the most "addictive" oncoprotein in many, if not all, such diseases. As a consequence of this tumor-specific dependency for MYB, it has become a major focus of efforts to develop specific antileukemia drugs. Much attention is being given to ways to interrupt the interaction between MYB and cooperating factors, in particular EP300/KAT3B and CBP/KAT3A. Aside from candidates identified through screening of small molecules, the most exciting prospect for novel drugs seems to be the design of peptide mimetics that interfere directly at the interface between MYB and its cofactors. Such peptides combine a high degree of target specificity with good efficacy including minimal effects on normal hematopoietic cells.
Collapse
Affiliation(s)
- Mary Louise Clarke
- Department of Biomedical Sciences, College of Medicine & Health, University of Birmingham, Edgbaston, Birmingham, UK
- Department of Cancer & Genomic Sciences, College of Medicine & Health, University of Birmingham, Edgbaston, Birmingham, UK
| | | | - Jon Frampton
- Department of Cancer & Genomic Sciences, College of Medicine & Health, University of Birmingham, Edgbaston, Birmingham, UK.
| |
Collapse
|
14
|
Boucher JC, Shrestha B, Vishwasrao P, Leick M, Cervantes EV, Ghafoor T, Reid K, Spitler K, Yu B, Betts BC, Guevara-Patino JA, Maus MV, Davila ML. Bispecific CD33/CD123 targeted chimeric antigen receptor T cells for the treatment of acute myeloid leukemia. Mol Ther Oncolytics 2023; 31:100751. [PMID: 38075241 PMCID: PMC10701585 DOI: 10.1016/j.omto.2023.100751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/16/2023] [Indexed: 02/12/2024] Open
Abstract
CD33 and CD123 are expressed on the surface of human acute myeloid leukemia blasts and other noncancerous tissues such as hematopoietic stem cells. On-target off-tumor toxicities may limit chimeric antigen receptor T cell therapies that target both CD33 and CD123. To overcome this limitation, we developed bispecific human CD33/CD123 chimeric antigen receptor (CAR) T cells with an "AND" logic gate. We produced novel CD33 and CD123 scFvs from monoclonal antibodies that bound CD33 and CD123 and activated T cells. Screening of CD33 and CD123 CAR T cells for cytotoxicity, cytokine production, and proliferation was performed, and we selected scFvs for CD33/CD123 bispecific CARs. The bispecific CARs split 4-1BB co-stimulation on one scFv and CD3ζ on the other. In vitro testing of cytokine secretion and cytotoxicity resulted in selecting bispecific CAR 1 construct for in vivo analysis. The CD33/CD123 bispecific CAR T cells were able to control acute myeloid leukemia (AML) in a xenograft AML mouse model similar to monospecific CD33 and CD123 CAR T cells while showing no on-target off-tumor effects. Based on our findings, human CD33/CD123 bispecific CAR T cells are a promising cell-based approach to prevent AML and support clinical investigation.
Collapse
Affiliation(s)
- Justin C. Boucher
- Department of Blood & Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Bishwas Shrestha
- Department of Blood & Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Paresh Vishwasrao
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, CA 91010, USA
- Department of Hematology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Mark Leick
- Cellular Immunotherapy Program. Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | | | | | - Kayla Reid
- Department of Blood & Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kristen Spitler
- Department of Blood & Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Bin Yu
- Department of Blood & Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Brian C. Betts
- Division of Hematology, Oncology, and Transplant, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Marcela V. Maus
- Cellular Immunotherapy Program. Massachusetts General Hospital Cancer Center, Boston, MA 02114, USA
| | - Marco L. Davila
- Department of Blood & Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Medicine and Immunology, Roswell Park Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
15
|
Pérez-Amill L, Bataller À, Delgado J, Esteve J, Juan M, Klein-González N. Advancing CART therapy for acute myeloid leukemia: recent breakthroughs and strategies for future development. Front Immunol 2023; 14:1260470. [PMID: 38098489 PMCID: PMC10720337 DOI: 10.3389/fimmu.2023.1260470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Chimeric antigen receptor (CAR) T therapies are being developed for acute myeloid leukemia (AML) on the basis of the results obtained for other haematological malignancies and the need of new treatments for relapsed and refractory AML. The biggest challenge of CART therapy for AML is to identify a specific target antigen, since antigens expressed in AML cells are usually shared with healthy haematopoietic stem cells (HSC). The concomitant expression of the target antigen on both tumour and HSC may lead to on-target/off-tumour toxicity. In this review, we guide researchers to design, develop, and translate to the clinic CART therapies for the treatment of AML. Specifically, we describe what issues have to be considered to design these therapies; what in vitro and in vivo assays can be used to prove their efficacy and safety; and what expertise and facilities are needed to treat and manage patients at the hospital.
Collapse
Affiliation(s)
- Lorena Pérez-Amill
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Gyala Therapeutics S.L, Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Àlex Bataller
- Department of Haematology, Institut Clínic de Malalties Hematològiques i Oncològiques (ICHMO), Hospital Clínic de Barcelona, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Julio Delgado
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Haematology, Institut Clínic de Malalties Hematològiques i Oncològiques (ICHMO), Hospital Clínic de Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Jordi Esteve
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Haematology, Institut Clínic de Malalties Hematològiques i Oncològiques (ICHMO), Hospital Clínic de Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Manel Juan
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
- Hospital Sant Joan de Déu, Universidad de Barcelona, Barcelona, Spain
| | - Nela Klein-González
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Gyala Therapeutics S.L, Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Garg S, Ni W, Griffin JD, Sattler M. Chimeric Antigen Receptor T Cell Therapy in Acute Myeloid Leukemia: Trials and Tribulations. Hematol Rep 2023; 15:608-626. [PMID: 37987319 PMCID: PMC10660693 DOI: 10.3390/hematolrep15040063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that is often associated with relapse and drug resistance after standard chemotherapy or targeted therapy, particularly in older patients. Hematopoietic stem cell transplants are looked upon as the ultimate salvage option with curative intent. Adoptive cell therapy using chimeric antigen receptors (CAR) has shown promise in B cell malignancies and is now being investigated in AML. Initial clinical trials have been disappointing in AML, and we review current strategies to improve efficacy for CAR approaches. The extensive number of clinical trials targeting different antigens likely reflects the genetic heterogeneity of AML. The limited number of patients reported in multiple early clinical studies makes it difficult to draw conclusions about CAR safety, but it does suggest that the efficacy of this approach in AML lags behind the success observed in B cell malignancies. There is a clear need not only to improve CAR design but also to identify targets in AML that show limited expression in normal myeloid lineage cells.
Collapse
Affiliation(s)
- Swati Garg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Ni
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - James D. Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Yang Z, Wang Y. Clinical development of chimeric antigen receptor-T cell therapy for hematological malignancies. Chin Med J (Engl) 2023; 136:2285-2296. [PMID: 37358555 PMCID: PMC10538902 DOI: 10.1097/cm9.0000000000002549] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Indexed: 06/27/2023] Open
Abstract
ABSTRACT Cellular therapies have revolutionized the treatment of hematological malignancies since their conception and rapid development. Chimeric antigen receptor (CAR)-T cell therapy is the most widely applied cellular therapy. Since the Food and Drug Administration approved two CD19-CAR-T products for clinical treatment of relapsed/refractory acute lymphoblastic leukemia and diffuse large B cell lymphoma in 2017, five more CAR-T cell products were subsequently approved for treating multiple myeloma or B cell malignancies. Moreover, clinical trials of CAR-T cell therapy for treating other hematological malignancies are ongoing. Both China and the United States have contributed significantly to the development of clinical trials. However, CAR-T cell therapy has many limitations such as a high relapse rate, adverse side effects, and restricted availability. Various methods are being implemented in clinical trials to address these issues, some of which have demonstrated promising breakthroughs. This review summarizes developments in CAR-T cell trials and advances in CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhihuan Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Tianjin Key Laboratory of Cell Therapy for Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | | |
Collapse
|
18
|
Almaeen AH, Abouelkheir M. CAR T-Cells in Acute Lymphoblastic Leukemia: Current Status and Future Prospects. Biomedicines 2023; 11:2693. [PMID: 37893067 PMCID: PMC10604728 DOI: 10.3390/biomedicines11102693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
The currently available treatment for acute lymphoblastic leukemia (ALL) is mainly dependent on the combination of chemotherapy, steroids, and allogeneic stem cell transplantation. However, refractoriness and relapse (R/R) after initial complete remission may reach up to 20% in pediatrics. This percentage may even reach 60% in adults. To overcome R/R, a new therapeutic approach was developed using what is called chimeric antigen receptor-modified (CAR) T-cell therapy. The Food and Drug Administration (FDA) in the United States has so far approved four CAR T-cells for the treatment of ALL. Using this new therapeutic strategy has shown a remarkable success in treating R/R ALL. However, the use of CAR T-cells is expensive, has many imitations, and is associated with some adverse effects. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are two common examples of these adverse effects. Moreover, R/R to CAR T-cell therapy can take place during treatment. Continuous development of this therapeutic strategy is ongoing to overcome these limitations and adverse effects. The present article overviews the use of CAR T-cell in the treatment of ALL, summarizing the results of relevant clinical trials and discussing future prospects intended to improve the efficacy of this therapeutic strategy and overcome its limitations.
Collapse
Affiliation(s)
- Abdulrahman H. Almaeen
- Department of Pathology, Pathology Division, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Mohamed Abouelkheir
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
19
|
Apavaloaei A, Perreault C. Immunotargeting of a recurrent AML-specific neoantigen. NATURE CANCER 2023; 4:1403-1405. [PMID: 37783806 DOI: 10.1038/s43018-023-00634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
|
20
|
Riva C, Vernarecci C, Minetto P, Goda R, Greppi M, Pesce S, Chies M, Zecchetti G, Ferro B, Maio E, Cea M, Lemoli RM, Marcenaro E, Guolo F. Harnessing Immune Response in Acute Myeloid Leukemia. J Clin Med 2023; 12:5824. [PMID: 37762763 PMCID: PMC10532363 DOI: 10.3390/jcm12185824] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Despite the results achieved with the evolution of conventional chemotherapy and the inclusion of targeted therapies in the treatment of acute myeloid leukemia (AML), survival is still not satisfying, in particular in the setting of relapsed/refractory (R/R) disease or elderly/unfit patients. Among the most innovative therapeutic options, cellular therapy has shown great results in different hematological malignancies such as acute lymphoblastic leukemia and lymphomas, with several products already approved for clinical use. However, despite the great interest in also expanding the application of these new treatments to R/R AML, no product has been approved yet for clinical application. Furthermore, cellular therapy could indeed represent a powerful tool and an appealing alternative to allogeneic hematopoietic stem cell transplantation for ineligible patients. In this review, we aim to provide an overview of the most recent clinical research exploring the effectiveness of cellular therapy in AML, moving from consolidated approaches such as post- transplant donor's lymphocytes infusion, to modern adoptive immunotherapies such as alloreactive NK cell infusions, engineered T and NK cells (CAR-T, CAR-NK) and novel platforms of T and NK cells engaging (i.e., BiTEs, DARTs and ANKETTM).
Collapse
Affiliation(s)
- Carola Riva
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Chiara Vernarecci
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Paola Minetto
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Rayan Goda
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Marco Greppi
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Silvia Pesce
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Maria Chies
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Giada Zecchetti
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Beatrice Ferro
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Elena Maio
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
| | - Michele Cea
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Roberto Massimo Lemoli
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Emanuela Marcenaro
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (R.G.); (M.G.); (S.P.)
| | - Fabio Guolo
- Clinic of Hematology, Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (C.R.); (C.V.); (M.C.); (G.Z.); (B.F.); (E.M.); (M.C.); (R.M.L.); (F.G.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| |
Collapse
|
21
|
Tosic N, Marjanovic I, Lazic J. Pediatric acute myeloid leukemia: Insight into genetic landscape and novel targeted approaches. Biochem Pharmacol 2023; 215:115705. [PMID: 37532055 DOI: 10.1016/j.bcp.2023.115705] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Acute myeloid leukemia (AML) is a very heterogeneous hematological malignancy that accounts for approximately 20% of all pediatric leukemia cases. The outcome of pediatric AML has improved over the last decades, with overall survival rates reaching up to 70%. Still, AML is among the leading types of pediatric cancers by its high mortality rate. Modulation of standard therapy, like chemotherapy intensification, hematopoietic stem cell transplantation and optimized supportive care, could only get this far, but for the significant improvement of the outcome in pediatric AML, development of novel targeted therapy approaches is necessary. In recent years the advances in genomic techniques have greatly expanded our knowledge of the AML biology, revealing molecular landscape and complexity of the disease, which in turn have led to the identification of novel therapeutic targets. This review provides a brief overview of the genetic landscape of pediatric AML, and how it's used for precise molecular characterization and risk stratification of the patients, and also for the development of effective targeted therapy. Furthermore, this review presents recent advances in molecular targeted therapy and immunotherapy with an emphasis on the therapeutic approaches with significant clinical benefits for pediatric AML.
Collapse
Affiliation(s)
- Natasa Tosic
- Institute of Molecular Genetics and Genetic Engineering, Laboratory for Molecular Biomedicine, University of Belgrade, Serbia.
| | - Irena Marjanovic
- Institute of Molecular Genetics and Genetic Engineering, Laboratory for Molecular Biomedicine, University of Belgrade, Serbia
| | - Jelena Lazic
- University Children's Hospital, Department for Hematology and Oncology, Belgrade, Serbia; Faculty of Medicine, University of Belgrade, Serbia
| |
Collapse
|
22
|
Casirati G, Cosentino A, Mucci A, Salah Mahmoud M, Ugarte Zabala I, Zeng J, Ficarro SB, Klatt D, Brendel C, Rambaldi A, Ritz J, Marto JA, Pellin D, Bauer DE, Armstrong SA, Genovese P. Epitope editing enables targeted immunotherapy of acute myeloid leukaemia. Nature 2023; 621:404-414. [PMID: 37648862 PMCID: PMC10499609 DOI: 10.1038/s41586-023-06496-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 07/28/2023] [Indexed: 09/01/2023]
Abstract
Despite the considerable efficacy observed when targeting a dispensable lineage antigen, such as CD19 in B cell acute lymphoblastic leukaemia1,2, the broader applicability of adoptive immunotherapies is hampered by the absence of tumour-restricted antigens3-5. Acute myeloid leukaemia immunotherapies target genes expressed by haematopoietic stem/progenitor cells (HSPCs) or differentiated myeloid cells, resulting in intolerable on-target/off-tumour toxicity. Here we show that epitope engineering of donor HSPCs used for bone marrow transplantation endows haematopoietic lineages with selective resistance to chimeric antigen receptor (CAR) T cells or monoclonal antibodies, without affecting protein function or regulation. This strategy enables the targeting of genes that are essential for leukaemia survival regardless of shared expression on HSPCs, reducing the risk of tumour immune escape. By performing epitope mapping and library screenings, we identified amino acid changes that abrogate the binding of therapeutic monoclonal antibodies targeting FLT3, CD123 and KIT, and optimized a base-editing approach to introduce them into CD34+ HSPCs, which retain long-term engraftment and multilineage differentiation ability. After CAR T cell treatment, we confirmed resistance of epitope-edited haematopoiesis and concomitant eradication of patient-derived acute myeloid leukaemia xenografts. Furthermore, we show that multiplex epitope engineering of HSPCs is feasible and enables more effective immunotherapies against multiple targets without incurring overlapping off-tumour toxicities. We envision that this approach will provide opportunities to treat relapsed/refractory acute myeloid leukaemia and enable safer non-genotoxic conditioning.
Collapse
Affiliation(s)
- Gabriele Casirati
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Milano-Bicocca University, Milan, Italy
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Andrea Cosentino
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- Department of Oncology and Hematology, University of Milan and Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Adele Mucci
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Mohammed Salah Mahmoud
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- Zoology Department, Faculty of Science, Fayoum University, Fayoum, Egypt
| | - Iratxe Ugarte Zabala
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jing Zeng
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Scott B Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Denise Klatt
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
| | - Christian Brendel
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- Harvard Medical School, Boston, MA, USA
| | - Alessandro Rambaldi
- Department of Oncology and Hematology, University of Milan and Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Jerome Ritz
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber Cancer Institute, Boston, USA
| | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Danilo Pellin
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- Harvard Medical School, Boston, MA, USA
| | - Scott A Armstrong
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA
- Harvard Medical School, Boston, MA, USA
| | - Pietro Genovese
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Atilla E, Benabdellah K. The Black Hole: CAR T Cell Therapy in AML. Cancers (Basel) 2023; 15:2713. [PMID: 37345050 DOI: 10.3390/cancers15102713] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Despite exhaustive studies, researchers have made little progress in the field of adoptive cellular therapies for relapsed/refractory acute myeloid leukemia (AML), unlike the notable uptake for B cell malignancies. Various single antigen-targeting chimeric antigen receptor (CAR) T cell Phase I trials have been established worldwide and have recruited approximately 100 patients. The high heterogeneity at the genetic and molecular levels within and between AML patients resembles a black hole: a great gravitational field that sucks in everything. One must consider the fact that only around 30% of patients show a response; there are, however, consequential off-tumor effects. It is obvious that a new point of view is needed to achieve more promising results. This review first introduces the unique therapeutic challenges of not only CAR T cells but also other adoptive cellular therapies in AML. Next, recent single-cell sequencing data for AML to assess somatically acquired alterations at the DNA, epigenetic, RNA, and protein levels are discussed to give a perspective on cellular heterogeneity, intercellular hierarchies, and the cellular ecosystem. Finally, promising novel strategies are summarized, including more sophisticated next-generation CAR T, TCR-T, and CAR NK therapies; the approaches with which to tailor the microenvironment and target neoantigens; and allogeneic approaches.
Collapse
Affiliation(s)
- Erden Atilla
- Fred Hutchinson Cancer Research Center, Clinical Research Division, 1100 Fairview Ave N, Seattle, WA 98109, USA
- GENYO Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer/University of Granada/Andalusian Regional Government, Health Sciences Technology Park, 18016 Granada, Spain
| | - Karim Benabdellah
- GENYO Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer/University of Granada/Andalusian Regional Government, Health Sciences Technology Park, 18016 Granada, Spain
| |
Collapse
|
24
|
Cheng J, Ge T, Zhu X, Wang J, Zeng Y, Mu W, Cai H, Dai Z, Jin J, Yang Y, Hu G, Mao X, Zhou J, Zhu L, Huang L. Preclinical development and evaluation of nanobody-based CD70-specific CAR T cells for the treatment of acute myeloid leukemia. Cancer Immunol Immunother 2023:10.1007/s00262-023-03422-6. [PMID: 36932256 DOI: 10.1007/s00262-023-03422-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/05/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) treatment remains challenging. CD70 was reported as a promising AML-specific antigen. Preclinically, CAR T-cell with single-chain-variable fragment (scFv) or truncated CD27 targeting CD70 has been reported to treat AML. However, various disadvantages including spontaneous exhaustion, proteinase-mediated loss of functional receptors, and high immunogenicity, limited its further application to clinical settings. Alternatively, the single-variable domain on heavy chain (VHH), also known as nanobodies, with comparable binding ability and specificity, provides an optional solution. METHOD We generated CD70 knocked-out novel nanobody-based anti-CD70-CAR T-cells (nb70CAR-T) with two different VHHs for antigen detection. Next, we detected the CD70 expression on primary AML blasts by flow cytometry and associated the efficacy of nb70CAR-T with the target antigen density. Finally, epigenetic modulators were investigated to regulate the CD70 expression on AML cells to promote the functionality of nb70CAR-T. RESULTS Our nb70CAR-T exhibited expected tumoricidal functionality against CD70-expressed cell lines and primary AML blasts. However, CD70 expression in primary AML blasts was not consistently high and nb70CAR-T potently respond to an estimated 40.4% of AML patients when the CD70 expression level was over a threshold of 1.6 (MFI ratio). Epigenetic modulators, Decitabine and Chidamide can up-regulate CD70 expression on AML cells, enhancing the treatment efficacy of nb70CAR-T. CONCLUSION CD70 expression in AML blasts was not fully supportive of its role in AML targeted therapy as reported. The combinational use of Chidamide and Decitabine with nb70CAR-T could provide a new potential for the treatment of AML.
Collapse
Affiliation(s)
- Jiali Cheng
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Tong Ge
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Jue Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Yuhao Zeng
- Department of Internal Medicine, Cleveland Clinic, Akron General, Akron, OH, USA
| | - Wei Mu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Haodong Cai
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Zhenyu Dai
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Jin Jin
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | | | - Guang Hu
- IASO Biotherapeutics, Nanjing, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Li Zhu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China.
| | - Liang Huang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China.
| |
Collapse
|
25
|
Teixeira A, Carreira L, Abalde-Cela S, Sampaio-Marques B, Areias AC, Ludovico P, Diéguez L. Current and Emerging Techniques for Diagnosis and MRD Detection in AML: A Comprehensive Narrative Review. Cancers (Basel) 2023; 15:cancers15051362. [PMID: 36900154 PMCID: PMC10000116 DOI: 10.3390/cancers15051362] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a group of hematologic neoplasms characterized by abnormal differentiation and proliferation of myeloid progenitor cells. AML is associated with poor outcome due to the lack of efficient therapies and early diagnostic tools. The current gold standard diagnostic tools are based on bone marrow biopsy. These biopsies, apart from being very invasive, painful, and costly, have low sensitivity. Despite the progress uncovering the molecular pathogenesis of AML, the development of novel detection strategies is still poorly explored. This is particularly important for patients that check the criteria for complete remission after treatment, since they can relapse through the persistence of some leukemic stem cells. This condition, recently named as measurable residual disease (MRD), has severe consequences for disease progression. Hence, an early and accurate diagnosis of MRD would allow an appropriate therapy to be tailored, improving a patient's prognosis. Many novel techniques with high potential in disease prevention and early detection are being explored. Among them, microfluidics has flourished in recent years due to its ability at processing complex samples as well as its demonstrated capacity to isolate rare cells from biological fluids. In parallel, surface-enhanced Raman scattering (SERS) spectroscopy has shown outstanding sensitivity and capability for multiplex quantitative detection of disease biomarkers. Together, these technologies can allow early and cost-effective disease detection as well as contribute to monitoring the efficiency of treatments. In this review, we aim to provide a comprehensive overview of AML disease, the conventional techniques currently used for its diagnosis, classification (recently updated in September 2022), and treatment selection, and we also aim to present how novel technologies can be applied to improve the detection and monitoring of MRD.
Collapse
Affiliation(s)
- Alexandra Teixeira
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Luís Carreira
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
| | - Sara Abalde-Cela
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
| | - Belém Sampaio-Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Anabela C. Areias
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Paula Ludovico
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s–PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (P.L.); (L.D.)
| | - Lorena Diéguez
- International Iberian Nanotechnology Laboratory (INL), Avda Mestre José Veiga, 4715-310 Braga, Portugal
- Correspondence: (P.L.); (L.D.)
| |
Collapse
|
26
|
Zhang H, Bu C, Peng Z, Li G, Zhou Z, Ding W, Zheng Y, He Y, Hu Z, Pei K, Luo M, Li C. Characteristics of anti-CLL1 based CAR-T therapy for children with relapsed or refractory acute myeloid leukemia: the multi-center efficacy and safety interim analysis. Leukemia 2022; 36:2596-2604. [PMID: 36151140 DOI: 10.1038/s41375-022-01703-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022]
Abstract
C-type lectin-like molecule-1 (CLL1) is preferentially expressed on acute myeloid leukemia (AML) stem cells and AML blasts, which can be considered as AML-associated antigen. Anti-CLL1-based CAR-T cells exhibited effective tumor-killing capacity in vitro and in AML-bearing mouse model. In this report, eight children with relapsed or refractory AML (R/R-AML) were recruited for a phase 1/2 clinical trial of autologous anti-CLL1 CAR-T cell immunotherapy. The objectives of this clinical trial were to evaluate the safety and the preliminary efficacy of anti-CLL1 CAR-T cell treatment. Patients received one dose of autologous anti-CLL1 CAR-T cells after lymphodepletion conditioning. After CAR-T treatment, patients developed grade 1-2 cytokine release syndrome (CRS) but without any lethal events. 4 out of 8 patients achieved morphologic leukemia-free state (MLFS) and minimal residual disease (MRD) negativity, 1 patient with MLFS and MRD positivity, 1 patient achieved complete remission with incomplete hematologic recovery (CRi) but MRD positivity, 1 patient with partial remission (PR), and 1 patient remained at stable disease (SD) status but had CLL1-positive AML blast clearance. These results suggested that anti-CLL1-based CAR-T cell immunotherapy can be considered as a well-tolerated and effective option for treating children with R/R-AML.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China.,Department of Hematology/Oncology, Shanghai Jiaotong University School of Medicine Affiliated Shanghai Children's Medical Center, Shanghai, China
| | - Chaoke Bu
- Nanfang-Chunfu Children's Institute of Hematology and Oncology, Taixin Hospital, Dongguan, China
| | - Zhiyong Peng
- Nanfang-Chunfu Children's Institute of Hematology and Oncology, Taixin Hospital, Dongguan, China
| | - Guangchao Li
- Guangzhou Bio-Gene Technology Co., Ltd, Guangzhou, China
| | - Zhao Zhou
- Guangzhou Bio-Gene Technology Co., Ltd, Guangzhou, China
| | - Wen Ding
- Guangzhou Bio-Gene Technology Co., Ltd, Guangzhou, China
| | - Yongwei Zheng
- Guangzhou Bio-Gene Technology Co., Ltd, Guangzhou, China
| | - Yingyi He
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Zhengbin Hu
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Kunlin Pei
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Min Luo
- Guangzhou Bio-Gene Technology Co., Ltd, Guangzhou, China.
| | - Chunfu Li
- Nanfang-Chunfu Children's Institute of Hematology and Oncology, Taixin Hospital, Dongguan, China.
| |
Collapse
|
27
|
Nogami A, Sasaki K. Therapeutic Advances in Immunotherapies for Hematological Malignancies. Int J Mol Sci 2022; 23:11526. [PMID: 36232824 PMCID: PMC9569660 DOI: 10.3390/ijms231911526] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
Following the success of immunotherapies such as chimeric antigen receptor transgenic T-cell (CAR-T) therapy, bispecific T-cell engager therapy, and immune checkpoint inhibitors in the treatment of hematologic malignancies, further studies are underway to improve the efficacy of these immunotherapies and to reduce the complications associated with their use in combination with other immune checkpoint inhibitors and conventional chemotherapy. Studies of novel therapeutic strategies such as bispecific (tandem or dual) CAR-T, bispecific killer cell engager, trispecific killer cell engager, and dual affinity retargeting therapies are also underway. Because of these studies and the discovery of novel immunotherapeutic target molecules, the use of immunotherapy for diseases initially thought to be less promising to treat with this treatment method, such as acute myeloid leukemia and T-cell hematologic tumors, has become a reality. Thus, in this coming era of new transplantation- and chemotherapy-free treatment strategies, it is imperative for both scientists and clinicians to understand the molecular immunity of hematologic malignancies. In this review, we focus on the remarkable development of immunotherapies that could change the prognosis of hematologic diseases. We also review the molecular mechanisms, development processes, clinical efficacies, and problems of new agents.
Collapse
Affiliation(s)
- Ayako Nogami
- Department of Laboratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
- Department of Hematology, Tokyo Medical and Dental University Hospital, 1-5-45 Yushima, Bunkyoku, Tokyo 1138510, Japan
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX 77030, USA
| |
Collapse
|
28
|
Huerga-Domínguez S, Villar S, Prósper F, Alfonso-Piérola A. Updates on the Management of Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:4756. [PMID: 36230677 PMCID: PMC9563665 DOI: 10.3390/cancers14194756] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/18/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Acute myeloid leukemia is a heterogeneous disease defined by a large spectrum of genetic aberrations that are potential therapeutic targets. New targeted therapies have changed the landscape for a disease with poor outcomes. They are more effective than standard chemotherapy with a good safety profile. For "fit patients" in first-line, the combination of gemtuzumab ozogamicin or midostaurin with intensive chemotherapy or Vyxeos is now considered the "standard of care" for selected patients. On the other hand, for "unfit patients", azacitidine-venetoclax has been consolidated as a frontline treatment, while other combinations with magrolimab or ivosidenib are in development. Nevertheless, global survival results, especially in relapsed or refractory patients, remain unfavorable. New immunotherapies or targeted therapies, such as Menin inhibitors or sabatolimab, represent an opportunity in this situation. Future directions will probably come from combinations of different targeted therapies ("triplets") and maintenance strategies guided by measurable residual disease.
Collapse
Affiliation(s)
| | | | | | - Ana Alfonso-Piérola
- Hematology and Hemotherapy Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
29
|
Vandghanooni S, Eskandani M, Sanaat Z, Omidi Y. Recent advances in the production, reprogramming, and application of CAR-T cells for treating hematological malignancies. Life Sci 2022; 309:121016. [PMID: 36179813 DOI: 10.1016/j.lfs.2022.121016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 10/31/2022]
Abstract
As genetically engineered cells, chimeric antigen receptor (CAR)-T cells express specific receptors on their surface to target and eliminate malignant cells. CAR proteins are equipped with elements that enhance the activity and survival of T cells. Once injected, CAR-T cells act as a "living drug" against tumor cells in the body. Up to now, CAR-T cell therapy has been demonstrated as a robust adoptive cell transfer (ACT) immunotherapeutic modality for eliminating tumor cells in refractory hematological malignancies. CAR-T cell therapy modality involves several steps, including the collecting of the blood from patients, the isolation of peripheral blood mononuclear cells (PBMCs), the enrichment of CD4+/CD8+ T cell, the genetic reprogramming, the expansion of modified T cells, and the injection of genetically engineered T cells. The production of CAR-T cells is a multi-step procedure, which needs precise and safety management systems, including good manufacturing practice (GMP), and in-line quality control and assurance. The current study describes the structure of CARs and concentrates on the next generations of CARs that are engaged in enhancing the anti-tumor responses and safety of the engineered T cells. This paper also highlights the important concerns in quality control and nonclinical research of CAR-T cells, as well as general insights into the manufacture, reprogramming, and application of CAR-T cells based on new and enhanced techniques for treating hematological malignancies. Besides, the application of the CRISPR-Cas9 genome editing technology and nanocarrier-based delivery systems containing CAR coding sequences to overcome the limitations of CAR-T cell therapy has also been explained.
Collapse
Affiliation(s)
- Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Sanaat
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
30
|
Trad R, Warda W, Alcazer V, Neto da Rocha M, Berceanu A, Nicod C, Haderbache R, Roussel X, Desbrosses Y, Daguindau E, Renosi F, Roumier C, Bouquet L, Biichle S, Guiot M, Seffar E, Caillot D, Depil S, Robinet E, Salma Y, Deconinck E, Deschamps M, Ferrand C. Chimeric antigen receptor T-cells targeting IL-1RAP: a promising new cellular immunotherapy to treat acute myeloid leukemia. J Immunother Cancer 2022; 10:jitc-2021-004222. [PMID: 35803613 PMCID: PMC9272123 DOI: 10.1136/jitc-2021-004222] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
Background Acute myeloid leukemia (AML) remains a very difficult disease to cure due to the persistence of leukemic stem cells (LSCs), which are resistant to different lines of chemotherapy and are the basis of refractory/relapsed (R/R) disease in 80% of patients with AML not receiving allogeneic transplantation. Methods In this study, we showed that the interleukin-1 receptor accessory protein (IL-1RAP) protein is overexpressed on the cell surface of LSCs in all subtypes of AML and confirmed it as an interesting and promising target in AML compared with the most common potential AML targets, since it is not expressed by the normal hematopoietic stem cell. After establishing the proof of concept for the efficacy of chimeric antigen receptor (CAR) T-cells targeting IL-1RAP in chronic myeloid leukemia, we hypothesized that third-generation IL-1RAP CAR T-cells could eliminate AML LSCs, where the medical need is not covered. Results We first demonstrated that IL-1RAP CAR T-cells can be produced from AML T-cells at the time of diagnosis and at relapse. In vitro and in vivo, we showed the effectiveness of IL-1RAP CAR T-cells against AML cell lines expressing different levels of IL-1RAP and the cytotoxicity of autologous IL-1RAP CAR T-cells against primary cells from patients with AML at diagnosis or at relapse. In patient-derived relapsed AML xenograft models, we confirmed that IL-1RAP CAR T-cells are able to circulate in peripheral blood and to migrate in the bone marrow and spleen, are cytotoxic against primary AML cells and increased overall survival. Conclusion In conclusion, our preclinical results suggest that IL-1RAP CAR T-based adoptive therapy could be a promising strategy in AML treatment and it warrants the clinical investigation of this CAR T-cell therapy.
Collapse
Affiliation(s)
- Rim Trad
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Walid Warda
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France.,CanCell Therapeutics, Besancon, France
| | | | - Mathieu Neto da Rocha
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France.,CanCell Therapeutics, Besancon, France
| | - Ana Berceanu
- Clinical Hematology, C.H. Univ Jean Minjoz, Besancon, France
| | | | | | - Xavier Roussel
- Clinical Hematology, C.H. Univ Jean Minjoz, Besancon, France
| | | | | | - Florain Renosi
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | | | - Lucie Bouquet
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Sabeha Biichle
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Melanie Guiot
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Evan Seffar
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France
| | - Denis Caillot
- Clinical Hematology, CHU François Mitterrand, Dijon, France
| | | | | | - Yahya Salma
- Laboratory of Applied Biotechnology (LBA3B), Lebanese University, Tripoli, Lebanon
| | - Eric Deconinck
- Clinical Hematology, C.H. Univ Jean Minjoz, Besancon, France
| | - Marina Deschamps
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France.,CanCell Therapeutics, Besancon, France
| | - Christophe Ferrand
- TIMC, EFSBFC, INSERM UMR1098 RIGHT,UFC, Besancon, France .,CanCell Therapeutics, Besancon, France
| |
Collapse
|
31
|
Koedam J, Wermke M, Ehninger A, Cartellieri M, Ehninger G. Chimeric antigen receptor T-cell therapy in acute myeloid leukemia. Curr Opin Hematol 2022; 29:74-83. [PMID: 35013048 PMCID: PMC8815830 DOI: 10.1097/moh.0000000000000703] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Treatment outcome of relapsed or refractory AML patients remains dismal and new treatment options are needed. Adoptive cell therapy using CAR-T cells is a potentially interesting approach in this. RECENT FINDINGS Several potentially interesting AML targets are being investigated with CAR-T therapy with over 60 clinical trials listed on clinicaltrials.gov. The first clinical data are only just emerging with mixed results, once more proving that further research is needed. SUMMARY Adoptive cell therapy using chimeric antigen receptor T cells is being investigated in AML through many clinical trials. So far, no AML-specific antigen has been identified, requiring additional strategies to mitigate on-target off-tumor toxicity and to increase efficacy. Focus point is to acquire control over the CAR T cells once administered. Strategies to do so include biodegradable CARs, inducible CARs, suicide-switch containing CARs and two-component modular CARs. Limited and mixed results are available, confirming the risk of lasting toxicity for nonswitchable CARs. Initial results of modular CARs suggest toxicity can be mitigated whilst maintaining CAR activity by the use of modular CAR concepts that allows for 'ON' and 'OFF' switching.
Collapse
Affiliation(s)
| | - Martin Wermke
- Division of Hematology, Oncology and Stem Cell Transplantation, Medical Clinic I, Department of Medicine I, University Hospital Carl Gustav Carus
- National Center for Tumor Diseases
| | | | | | | |
Collapse
|
32
|
Role of Biomarkers in FLT3 AML. Cancers (Basel) 2022; 14:cancers14051164. [PMID: 35267471 PMCID: PMC8909069 DOI: 10.3390/cancers14051164] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Genetically heterogeneous disorder acute myeloid leukemia (AML) is marked by recurring mutations in FLT3. Current FLT3 inhibitors and other emerging inhibitors have helped in the improvement of the quality of standard of care therapies; however, the overall survival of the patients remains static. This is due to numerous mutations in FLT3, which causes resistance against these FLT3 inhibitors. For effective treatment of AML patients, alternative approaches are required to overcome this resistance. Here, we will summarize the biomarkers for FLT3 inhibitors in AML, as well as the alternative measures to overcome resistance to the current therapies. Abstract Acute myeloid leukemia is a disease characterized by uncontrolled proliferation of clonal myeloid blast cells that are incapable of maturation to leukocytes. AML is the most common leukemia in adults and remains a highly fatal disease with a five-year survival rate of 24%. More than 50% of AML patients have mutations in the FLT3 gene, rendering FLT3 an attractive target for small-molecule inhibition. Currently, there are several FLT3 inhibitors in the clinic, and others remain in clinical trials. However, these inhibitors face challenges due to lack of efficacy against several FLT3 mutants. Therefore, the identification of biomarkers is vital to stratify AML patients and target AML patient population with a particular FLT3 mutation. Additionally, there is an unmet need to identify alternative approaches to combat the resistance to FLT3 inhibitors. Here, we summarize the current knowledge on the utilization of diagnostic, prognostic, predictive, and pharmacodynamic biomarkers for FLT3-mutated AML. The resistance mechanisms to various FLT3 inhibitors and alternative approaches to combat this resistance are also discussed and presented.
Collapse
|
33
|
[Development and functional verification of CAR-T cells targeting CLL-1]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:102-106. [PMID: 35381669 PMCID: PMC8980646 DOI: 10.3760/cma.j.issn.0253-2727.2022.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Objective: To explore the development of a CAR-T cells targeting CLL-1 and verify its function. Methods: The expression levels of CLL-1 targets in cell lines and primary cells were detected by flow cytometry. A CLL-1 CAR vector was constructed, and the corresponding lentivirus was prepared. After infection and activation of T cells, CAR-T cells targeting CLL-1 were produced and their function was verified in vitro and in vivo. Results: CLL-1 was expressed in acute myeloid leukemia (AML) cell lines and primary AML cells. The transduction rate of the prepared CAR T cells was 77.82%. In AML cell lines and AML primary cells, CLL-1-targeting CAR-T cells significantly and specifically killed CLL-1-expressing cells. Compared to untransduced T cells, CAR-T cells killed target cells and secreted inflammatory cytokines, such as interleukin-6 and interferon-γ, at significantly higher levels (P<0.001) . In an in vivo human xenograft mouse model of AML, CLL-1 CAR-T cells also exhibited potent antileukemic activity and induced prolonged mouse survival compared with untransduced T cells [not reached vs 22 days (95%CI 19-24 days) , P=0.002]. Conclusion: CAR-T cells targeting CLL-1 have been successfully produced and have excellent functions.
Collapse
|
34
|
Badar T, Manna A, Gadd ME, Kharfan-Dabaja MA, Qin H. Prospect of CAR T-cell therapy in acute myeloid leukemia. Expert Opin Investig Drugs 2022; 31:211-220. [PMID: 35051347 DOI: 10.1080/13543784.2022.2032642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Long-term outcome of patients with acute myeloid leukemia (AML) remains dismal, especially for those with high-risk disease or who are refractory to conventional therapy. CAR T-cell therapy provides unique opportunity to improve outcome by specifically targeting leukemia cells through genetically engineered T-cells. AREAS COVERED We summarize the progress of CAR T-cells therapy in AML. We examine its shortcomings in AML therapy and the strategies that are being implemented to improve its safety and effectiveness. PubMed Central, ClinicalTrials.gov and ASH annual meeting abstracts, were searched. Search terms used to identify clinical trials were "CAR T-cells in AML" OR CAR T-cells in leukemia". Relevant clinical trials and CAR T-cell research data was reviewed from June 2009 till July 2021. EXPERT OPINION CAR T-cell therapy has shown promise as a novel therapy, but there are number of barriers to overcome to achieve it full therapeutic potential in AML. Targeting leukemia specific antigen such as CLL1, to avoid myelotoxicity; incorporating checkpoint inhibitors to overcome leukemia induced immunosuppression and allogenic CAR T-cells to increases accessibility to patients with proliferative disease are among the strategies that are being explored to make CAR T-cell a successful immunotherapy for patient with AML.
Collapse
Affiliation(s)
- Talha Badar
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Florida, USA
| | - Alak Manna
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Florida, USA
| | - Martha E Gadd
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Florida, USA
| | | | - Hong Qin
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Florida, USA
| |
Collapse
|
35
|
Sheykhhasan M, Manoochehri H, Dama P. Use of CAR T-cell for acute lymphoblastic leukemia (ALL) treatment: a review study. Cancer Gene Ther 2022; 29:1080-1096. [PMID: 34987176 PMCID: PMC9395272 DOI: 10.1038/s41417-021-00418-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 11/18/2021] [Accepted: 12/13/2021] [Indexed: 02/08/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is a cancer-specific lymphoid cell. Induction and consolidation chemotherapy alone or in combination with different therapeutic approaches remain the main treatment. Although complete or partial remission of the disease can be achieved, the risk of relapse or refractory leukemia is still high. More effective and safe therapy options are yet unmet needs. In recent years' new therapeutic approaches have been widely used. Hematopoietic Stem Cell Transplantation (HSCT) presents significant limitations and the outcome of the consolidation treatment is patient dependent. Side effects such as Graft versus Host Disease (GvHD) in allogeneic hematopoietic stem cell transplantation are extremely common, therefore, using alternative methods to address these challenges for treatment seems crucial. In the last decade, T cells genetically engineered with Chimeric Antigen Receptor (CAR) treatment for the ALL are largely studied and represent the new era of strategy. According to the Phase I/II clinical trials, this technology results seem very promising and can be used in the next future as an effective and safe treatment for ALL treatment. In this review different generations, challenges, and clinical studies related to chimeric antigen receptor (CAR) T-cells for ALL treatment are discussed.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- grid.411950.80000 0004 0611 9280Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran ,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - Hamed Manoochehri
- grid.411950.80000 0004 0611 9280Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Paola Dama
- Research Fellow School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
36
|
Soltani M, Zhao Y, Xia Z, Ganjalikhani Hakemi M, Bazhin AV. The Importance of Cellular Metabolic Pathways in Pathogenesis and Selective Treatments of Hematological Malignancies. Front Oncol 2021; 11:767026. [PMID: 34868994 PMCID: PMC8636012 DOI: 10.3389/fonc.2021.767026] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/20/2021] [Indexed: 02/05/2023] Open
Abstract
Despite recent advancements in the treatment of hematologic malignancies and the emergence of newer and more sophisticated therapeutic approaches such as immunotherapy, long-term overall survival remains unsatisfactory. Metabolic alteration, as an important hallmark of cancer cells, not only contributes to the malignant transformation of cells, but also promotes tumor progression and metastasis. As an immune-escape mechanism, the metabolic adaptation of the bone marrow microenvironment and leukemic cells is a major player in the suppression of anti-leukemia immune responses. Therefore, metabolic rewiring in leukemia would provide promising opportunities for newer therapeutic interventions. Several therapeutic agents which affect essential bioenergetic pathways in cancer cells including glycolysis, β-oxidation of fatty acids and Krebs cycle, or anabolic pathways such as lipid biosynthesis and pentose phosphate pathway, are being tested in various types of cancers. So far, numerous preclinical or clinical trial studies using such metabolic agents alone or in combination with other remedies such as immunotherapy are in progress and have demonstrated promising outcomes. In this review, we aim to argue the importance of metabolic alterations and bioenergetic pathways in different types of leukemia and their vital roles in disease development. Designing treatments based on targeting leukemic cells vulnerabilities, particularly in nonresponsive leukemia patients, should be warranted.
Collapse
Affiliation(s)
- Mojdeh Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yue Zhao
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | | | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
37
|
Nian Z, Zheng X, Dou Y, Du X, Zhou L, Fu B, Sun R, Tian Z, Wei H. Rapamycin Pretreatment Rescues the Bone Marrow AML Cell Elimination Capacity of CAR-T Cells. Clin Cancer Res 2021; 27:6026-6038. [PMID: 34233960 PMCID: PMC9401534 DOI: 10.1158/1078-0432.ccr-21-0452] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/26/2021] [Accepted: 06/30/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Ongoing clinical trials show limited efficacy for Chimeric antigen receptor (CAR) T treatment for acute myeloid leukemia (AML). The aim of this study was to identify potential causes of the reported limited efficacy from CAR-T therapies against AML. EXPERIMENTAL DESIGN We generated CAR-T cells targeting Epithelial cell adhesion molecule (EpCAM) and evaluated their killing activity against AML cells. We examined the impacts of modulating mTORC1 and mTORC2 signaling in CAR-T cells in terms of CXCR4 levels. We examined the effects of a rapamycin pretreatment of EpCAM CAR-T cells (during ex vivo expansion) and assessed the in vivo antitumor efficacy of rapamycin-pretreated EpCAM CAR-T cells (including CXCR4 knockdown cells) and CD33 CAR-T cells in leukemia xenograft mouse models. RESULTS EpCAM CAR-T exhibited killing activity against AML cells but failed to eliminate AML cells in bone marrow. Subsequent investigations revealed that aberrantly activated mTORC1 signaling in CAR-T cells results in decreased bone marrow infiltration and decreased the levels of the rapamycin target CXCR4. Attenuating mTORC1 activity with the rapamycin pretreatment increased the capacity of CAR-T cells to infiltrate bone marrow and enhanced the extent of bone marrow AML cell elimination in leukemia xenograft mouse models. CXCR4 knockdown experiments showed that CXCR4 contributes to the enhanced bone marrow infiltration capacity of EpCAM CAR-T cells and the observed reduction in bone marrow AML cells. CONCLUSIONS Our study reveals a potential cause for the limited efficacy of CAR-T reported from current AML clinical trials and illustrates an easy-to-implement pretreatment strategy, which enhances the anti-AML efficacy of CAR-T cells.See related commentary by Maiti and Daver, p. 5739.
Collapse
Affiliation(s)
- Zhigang Nian
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiaohu Zheng
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Corresponding Authors: Haiming Wei, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui 230027, China. Phone: 0551-6360-7379; E-mail: ; and Xiaohu Zheng, E-mail:
| | - Yingchao Dou
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xianghui Du
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Li Zhou
- Department of Hematology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Binqing Fu
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Rui Sun
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China.,Corresponding Authors: Haiming Wei, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui 230027, China. Phone: 0551-6360-7379; E-mail: ; and Xiaohu Zheng, E-mail:
| |
Collapse
|
38
|
Kaleka G, Schiller G. Immunotherapy for Acute Myeloid Leukemia: Allogeneic hematopoietic cell transplantation is here to stay. Leuk Res 2021; 112:106732. [PMID: 34864447 DOI: 10.1016/j.leukres.2021.106732] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 01/20/2023]
Abstract
Acute Myeloid Leukemia (AML) represents 1 % of all new cancer diagnosis made annually in the US and has a five-year survival of 30 %. Traditional treatment includes aggressive induction therapy followed by consolidation therapy that may include a hematopoietic stem cell transplant (HSCT). Thus far, HSCT remains the only potentially curative therapy for many patients with AML owing to the graft-versus-leukemia effect elicited by this treatment. The use of novel therapies, specifically immunotherapy, in the treatment of AML has been limited by the lack of appropriate target antigens, therapy associated toxicities and variable success with treatment. Antigenic variability on leukemia cells and the sharing of antigens by malignant and non-malignant cells makes the identification of appropriate antigens problematic. While studies with immunotherapeutic agents are underway, prior investigations have demonstrated a mixed response with some studies prematurely discontinued due to associated toxicities. This review presents a discussion of the envisioned role of immunotherapy in the treatment of AML in the setting of mixed therapeutic success and potentially lethal toxicities.
Collapse
Affiliation(s)
- Guneet Kaleka
- UCLA-Olive View Medical Center, Department of Medicine, Room 2B-182, 14445 Olive View Drive, Sylmar, CA, 91342, United States.
| | - Gary Schiller
- Department of Medicine, Hematology & Oncology at UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
39
|
Goel H, Rahul E, Gupta I, Chopra A, Ranjan A, Gupta AK, Meena JP, Viswanathan GK, Bakhshi S, Misra A, Hussain S, Kumar R, Singh A, Rath GK, Sharma A, Mittan S, Tanwar P. Molecular and genomic landscapes in secondary & therapy related acute myeloid leukemia. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:472-497. [PMID: 34824881 PMCID: PMC8610791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/04/2021] [Indexed: 06/13/2023]
Abstract
Acute myeloid leukemia (AML) is a complex, aggressive myeloid neoplasm characterized by frequent somatic mutations that influence different functional categories' genes, resulting in maturational arrest and clonal expansion. AML can arise de novo (dn-AML) or can be secondary AML (s-AML) refers to a leukemic process which may arise from an antecedent hematologic disorder (AHD-AML), mostly from a myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN) or can be the result of an antecedent cytotoxic chemotherapy or radiation therapy (therapy-related AML, t-AML). Clinical and biological features in secondary and therapy-related AML are distinct from de novo AML. Secondary and therapy-related AML occurs mainly in the elderly population and responds worse to therapy with higher relapse rates due to resistance to cytotoxic chemotherapy. Over the last decade, advances in molecular genetics have disclosed the sub-clonal architecture of secondary and therapy-related AML. Recent investigations have revealed that cytogenetic abnormalities and underlying genetic aberrations (mutations) are likely to be significant factors dictating prognosis and critical impacts on treatment outcome. Secondary and therapy-related AML have a poorer outcome with adverse cytogenetic abnormalities and higher recurrences of unfavorable mutations compared to de novo AML. In this review, we present an overview of the clinical features of secondary and therapy-related AML and address the function of genetic mutations implicated in the pathogenesis of secondary leukemia. Detailed knowledge of the pathogenetic mechanisms gives an overview of new prognostic markers, including targetable mutations that will presumably lead to the designing and developing novel molecular targeted therapies for secondary and therapy-related AML. Despite significant advances in knowing the genetic aspect of secondary and therapy-related AML, its influence on the disease's pathophysiology, standard treatment prospects have not significantly evolved during the past three decades. Thus, we conclude this review by summarizing the modern and developing treatment strategies in secondary and therapy-related acute myeloid leukemia.
Collapse
Affiliation(s)
- Harsh Goel
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ekta Rahul
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Ishan Gupta
- All India Institute of Medical SciencesNew Delhi 110029, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Amar Ranjan
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| | - Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Ganesh Kumar Viswanathan
- Department of Hematology, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Sameer Bakhshi
- Department of Medical Oncology, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Aroonima Misra
- National Institute of Pathology, ICMRNew Delhi 110029, India
| | - Showket Hussain
- Division Of Molecular Oncology, National Institute of Cancer Prevention & Research I-7, Sector-39Noida 201301, India
| | - Ritesh Kumar
- Department of Radiation Oncology, Rudgers Cancer Institute of New JerseyNJ 07103, United States
| | - Archana Singh
- Department of Pathology, College of Medical Sciences, Rajasthan University of Health SciencesJaipur 302033, India
| | - GK Rath
- Department of Radiotherapy, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences New DelhiNew Delhi 110029, India
| | - Sandeep Mittan
- Department of Cardiology, Ichan School of Medicine, Mount Sinai Hospital1468 Madison Avenue, New York 10028, United States
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr.B.R.A. Institute Rotary Cancer Hospital All India Institute of Medical SciencesNew Delhi 110029, India
| |
Collapse
|
40
|
Abstract
Acute myeloid leukemia (AML) is an uncommon but potentially catastrophic diagnosis with historically high mortality rates. The standard of care treatment remained unchanged for decades; however, recent discoveries of molecular drivers of leukemogenesis and disease progression have led to novel therapies for AML. Ongoing research and clinical trials are actively seeking to personalize therapy by identifying molecular targets, discovering patient specific and disease specific risk factors, and identifying effective combinations of modalities and drugs. This review focuses on important updates in diagnostic and disease classifications that reflect new understanding of the biology of AML, its mutational heterogeneity, some important genetic and environmental risk factors, and new treatment options including cytotoxic chemotherapy, novel targeted agents, and cellular therapies.
Collapse
Affiliation(s)
- Laura F Newell
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR, USA
| | - Rachel J Cook
- Knight Cancer Institute, Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
41
|
Rennert PD, Dufort FJ, Su L, Sanford T, Birt A, Wu L, Lobb RR, Ambrose C. Anti-CD19 CAR T Cells That Secrete a Biparatopic Anti-CLEC12A Bridging Protein Have Potent Activity Against Highly Aggressive Acute Myeloid Leukemia In Vitro and In Vivo. Mol Cancer Ther 2021; 20:2071-2081. [PMID: 34253594 PMCID: PMC9398100 DOI: 10.1158/1535-7163.mct-20-1030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/02/2021] [Accepted: 07/02/2021] [Indexed: 01/07/2023]
Abstract
Refractory acute myeloid leukemia (AML) remains an incurable malignancy despite the clinical use of novel targeted therapies, new antibody-based therapies, and cellular therapeutics. Here, we describe the preclinical development of a novel cell therapy that targets the antigen CLEC12A with a biparatopic bridging protein. Bridging proteins are designed as "CAR-T cell engagers," with a CAR-targeted protein fused to antigen binding domains derived from antibodies. Here, we created a CD19-anti-CLEC12A bridging protein that binds to CAR19 T cells and to the antigen CLEC12A. Biparatopic targeting increases the potency of bridging protein-mediated cytotoxicity by CAR19 T cells. Using CAR19 T cells that secrete the bridging protein we demonstrate potent activity against aggressive leukemic cell lines in vivo This CAR-engager platform is facile and modular, as illustrated by activity of a dual-antigen bridging protein targeting CLEC12A and CD33, designed to counter tumor heterogeneity and antigen escape, and created without the need for extensive CAR T-cell genetic engineering. CAR19 T cells provide an optimal cell therapy platform with well-understood inherent persistence and fitness characteristics.
Collapse
Affiliation(s)
- Paul D. Rennert
- Corresponding Author: Paul D. Rennert, Research & Development, Aleta Biotherapeutics Inc., Natick, MA 01760. Phone: 508-282-6370; E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Trial Watch: Adoptive TCR-Engineered T-Cell Immunotherapy for Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13184519. [PMID: 34572745 PMCID: PMC8469736 DOI: 10.3390/cancers13184519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is a type of blood cancer with an extremely grim prognosis. This is due to the fact that the majority of patients will relapse after frontline treatment. Overall survival of relapsed AML is very low, and treatment options are few. T lymphocytes harnessed with antitumor T-cell receptors (TCRs) can produce objective clinical responses in certain cancers, such as melanoma, but have not entered the main road for AML. In this review, we describe the current status of the field of TCR-T-cell therapies for AML. Abstract Despite the advent of novel therapies, acute myeloid leukemia (AML) remains associated with a grim prognosis. This is exemplified by 5-year overall survival rates not exceeding 30%. Even with frontline high-intensity chemotherapy regimens and allogeneic hematopoietic stem cell transplantation, the majority of patients with AML will relapse. For these patients, treatment options are few, and novel therapies are urgently needed. Adoptive T-cell therapies represent an attractive therapeutic avenue due to the intrinsic ability of T lymphocytes to recognize tumor cells with high specificity and efficiency. In particular, T-cell therapies focused on introducing T-cell receptors (TCRs) against tumor antigens have achieved objective clinical responses in solid tumors such as synovial sarcoma and melanoma. However, contrary to chimeric antigen receptor (CAR)-T cells with groundbreaking results in B-cell malignancies, the use of TCR-T cells for hematological malignancies is still in its infancy. In this review, we provide an overview of the status and clinical advances in adoptive TCR-T-cell therapy for the treatment of AML.
Collapse
|
43
|
Gorin NC. History and Development of Autologous Stem Cell Transplantation for Acute Myeloid Leukemia. Clin Hematol Int 2021; 3:83-95. [PMID: 34820613 PMCID: PMC8486970 DOI: 10.2991/chi.k.210703.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/03/2021] [Indexed: 11/23/2022] Open
Abstract
This review describes the development of cryopreservation, the birth of autologous stem cell transplantation (ASCT) and its past and present use to consolidate adult patients with acute myelogenous leukemia (AML). It summarizes the first autografts in patients in relapse, the experience of autografting in complete remission (CR), using bone marrow unpurged or purged in vitro with cyclophosphamide-derivatives, and the important shift to peripheral blood stem cells. The review also discusses the results of recent studies in favor of the use of ASCT to consolidate good- and intermediate-risk patients who reach CR with no detectable minimal residual disease, and those which support the inclusion of maintenance therapy post autograft with hypomethylating agents, anti-BCL-2, and, possibly, in the future, anti AML chimeric antigen receptor-T cells. Carefully applied to well-selected patients, ASCT may regain interest, because of its simplicity, its reduced toxicity, lower non-relapse mortality and better quality of life.
Collapse
Affiliation(s)
- Norbert Claude Gorin
- Department of Hematology and Cell Therapy, and EBMT Global Committee, Hopital Saint-Antoine APHP, Paris Sorbonne University, Paris, France
| |
Collapse
|
44
|
Lu H, Zhao X, Li Z, Hu Y, Wang H. From CAR-T Cells to CAR-NK Cells: A Developing Immunotherapy Method for Hematological Malignancies. Front Oncol 2021; 11:720501. [PMID: 34422667 PMCID: PMC8377427 DOI: 10.3389/fonc.2021.720501] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022] Open
Abstract
The approval of CD19 chimeric antigen receptor (CAR)-engineered T (CAR-T) cell products in B-cell malignancies represents a breakthrough in CAR-T cell immunotherapy. However, the remaining limitations concerning the graft-versus-host disease (GVHD) and other adverse effects (e.g., cytokine release syndromes [CRS] and neurotoxicity) still restrict their wider applications. Natural killer (NK) cells have been identified as promising candidates for CAR-based cellular immunotherapy because of their unique characteristics. No HLA-matching restriction and abundant sources make CAR-engineered NK (CAR-NK) cells potentially available to be off-the-shelf products that could be readily available for immediate clinical use. Therefore, researchers have gradually shifted their focus from CAR-T cells to CAR-NK cells in hematological malignancies. This review discusses the current status and applications of CAR-NK cells in hematological malignancies, as well as the unique advantages of CAR-NK cells compared with CAR-T cells. It also discusses challenges and prospects regarding clinical applications of CAR-NK cells.
Collapse
Affiliation(s)
- Hui Lu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Zhao
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziying Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huafang Wang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Marofi F, Rahman HS, Al-Obaidi ZMJ, Jalil AT, Abdelbasset WK, Suksatan W, Dorofeev AE, Shomali N, Chartrand MS, Pathak Y, Hassanzadeh A, Baradaran B, Ahmadi M, Saeedi H, Tahmasebi S, Jarahian M. Novel CAR T therapy is a ray of hope in the treatment of seriously ill AML patients. Stem Cell Res Ther 2021; 12:465. [PMID: 34412685 PMCID: PMC8377882 DOI: 10.1186/s13287-021-02420-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is a serious, life-threatening, and hardly curable hematological malignancy that affects the myeloid cell progenies and challenges patients of all ages but mostly occurs in adults. Although several therapies are available including chemotherapy, allogeneic hematopoietic stem cell transplantation (alloHSCT), and receptor-antagonist drugs, the 5-year survival of patients is quietly disappointing, less than 30%. alloHSCT is the major curative approach for AML with promising results but the treatment has severe adverse effects such as graft-versus-host disease (GVHD). Therefore, as an alternative, more efficient and less harmful immunotherapy-based approaches such as the adoptive transferring T cell therapy are in development for the treatment of AML. As such, chimeric antigen receptor (CAR) T cells are engineered T cells which have been developed in recent years as a breakthrough in cancer therapy. Interestingly, CAR T cells are effective against both solid tumors and hematological cancers such as AML. Gradually, CAR T cell therapy found its way into cancer therapy and was widely used for the treatment of hematologic malignancies with successful results particularly with somewhat better results in hematological cancer in comparison to solid tumors. The AML is generally fatal, therapy-resistant, and sometimes refractory disease with a disappointing low survival rate and weak prognosis. The 5-year survival rate for AML is only about 30%. However, the survival rate seems to be age-dependent. Novel CAR T cell therapy is a light at the end of the tunnel. The CD19 is an important target antigen in AML and lymphoma and the CAR T cells are engineered to target the CD19. In addition, a lot of research goes on the discovery of novel target antigens with therapeutic efficacy and utilizable for generating CAR T cells against various types of cancers. In recent years, many pieces of research on screening and identification of novel AML antigen targets with the goal of generation of effective anti-cancer CAR T cells have led to new therapies with strong cytotoxicity against cancerous cells and impressive clinical outcomes. Also, more recently, an improved version of CAR T cells which were called modified or smartly reprogrammed CAR T cells has been designed with less unwelcome effects, less toxicity against normal cells, more safety, more specificity, longer persistence, and proliferation capability. The purpose of this review is to discuss and explain the most recent advances in CAR T cell-based therapies targeting AML antigens and review the results of preclinical and clinical trials. Moreover, we will criticize the clinical challenges, side effects, and the different strategies for CAR T cell therapy.
Collapse
Affiliation(s)
- Faroogh Marofi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Heshu Sulaiman Rahman
- College of Medicine, University of Sulaimani, Sulaimaniyah, Iraq.,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaise, Sulaimaniyah, Iraq
| | - Zaid Mahdi Jaber Al-Obaidi
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Alkafeel, Najaf, 54001, Iraq.,Department of Chemistry and Biochemistry, College of Medicine, University of Kerbala, Karbala, 56001, Iraq
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia.,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | | | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Yashwant Pathak
- Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA.,Department of Pharmaceutics, Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Ali Hassanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Safa Tahmasebi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy, No. 2, Floor 4 Unit (G401), 69120, Heidelberg, Germany.
| |
Collapse
|
46
|
Sauer T, Parikh K, Sharma S, Omer B, Sedloev D, Chen Q, Angenendt L, Schliemann C, Schmitt M, Müller-Tidow C, Gottschalk S, Rooney CM. CD70-specific CAR T cells have potent activity against acute myeloid leukemia without HSC toxicity. Blood 2021; 138:318-330. [PMID: 34323938 PMCID: PMC8323977 DOI: 10.1182/blood.2020008221] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
The prognosis of patients with acute myeloid leukemia (AML) remains dismal, highlighting the need for novel innovative treatment strategies. The application of chimeric antigen receptor (CAR) T-cell therapy to patients with AML has been limited, in particular by the lack of a tumor-specific target antigen. CD70 is a promising antigen to target AML, as it is expressed on most leukemic blasts, whereas little or no expression is detectable in normal bone marrow samples. To target CD70 on AML cells, we generated a panel of CD70-CAR T cells that contained a common single-chain variable fragment (scFv) for antigen detection, but differed in size and flexibility of the extracellular spacer and in the transmembrane and the costimulatory domains. These CD70scFv CAR T cells were compared with a CAR construct that contained human CD27, the ligand of CD70 fused to the CD3ζ chain (CD27z). The structural composition of the CAR strongly influenced expression levels, viability, expansion, and cytotoxic capacities of CD70scFv-based CAR T cells, but CD27z-CAR T cells demonstrated superior proliferation and antitumor activity in vitro and in vivo, compared with all CD70scFv-CAR T cells. Although CD70-CAR T cells recognized activated virus-specific T cells (VSTs) that expressed CD70, they did not prevent colony formation by normal hematopoietic stem cells. Thus, CD70-targeted immunotherapy is a promising new treatment strategy for patients with CD70-positive AML that does not affect normal hematopoiesis but will require monitoring of virus-specific T-cell responses.
Collapse
Affiliation(s)
- Tim Sauer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Kathan Parikh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| | - David Sedloev
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Qian Chen
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Linus Angenendt
- Department of Internal Medicine A, University Hospital of Muenster, Muenster, Germany; and
| | - Christoph Schliemann
- Department of Internal Medicine A, University Hospital of Muenster, Muenster, Germany; and
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| |
Collapse
|
47
|
Meyer JE, Loff S, Dietrich J, Spehr J, Jurado Jiménez G, von Bonin M, Ehninger G, Cartellieri M, Ehninger A. Evaluation of switch-mediated costimulation in trans on universal CAR-T cells (UniCAR) targeting CD123-positive AML. Oncoimmunology 2021; 10:1945804. [PMID: 34290907 PMCID: PMC8274446 DOI: 10.1080/2162402x.2021.1945804] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR-T) targeting CD19 have achieved significant success in patients with B cell malignancies. To date, implementation of CAR-T in other indications remains challenging due to the lack of truly tumor-specific antigens as well as control of CAR-T activity in patients. CD123 is highly expressed in acute myeloid leukemia (AML) blasts including leukemia-initiating cells making it an attractive immunotherapeutic target. However, CD123 expression in normal hematopoietic progenitor cells and endothelia bears the risk of severe toxicities and may limit CAR-T applications lacking fine-tuned control mechanisms. Therefore, we recently developed a rapidly switchable universal CAR-T platform (UniCAR), in which CAR-T activity depends on the presence of a soluble adapter called targeting module (TM), and confirmed clinical proof-of-concept for targeting CD123 in AML with improved safety. As costimulation via 4–1BB ligand (4–1BBL) can enhance CAR-T expansion, persistence, and effector functions, a novel CD123-specific TM variant (TM123-4-1BBL) comprising trimeric single-chain 4–1BBL was developed for transient costimulation of UniCAR-T cells (UniCAR-T) at the leukemic site in trans. TM123-4-1BBL-directed UniCAR-T efficiently eradicated CD123-positive AML cells in vitro and in a CDX in vivo model. Moreover, additional costimulation via TM123-4-1BBL enabled enhanced expansion and persistence with a modulated UniCAR-T phenotype. In addition, the increased hydrodynamic volume of TM123-4-1BBL prolonged terminal plasma half-life and ensured a high total drug exposure in vivo. In conclusion, expanding the soluble adapter optionality for CD123-directed UniCAR-T maintains the platforms high anti-leukemic efficacy and immediate control mechanism for a flexible, safe, and individualized CAR-T therapy of AML patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Malte von Bonin
- Division of Hematology, Oncology and Stem Cell Transplantation, Medical Clinic I, Department of Medicine I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | | | | |
Collapse
|
48
|
Immunotherapy in AML: a brief review on emerging strategies. Clin Transl Oncol 2021; 23:2431-2447. [PMID: 34160771 DOI: 10.1007/s12094-021-02662-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
Acute myeloid leukemia (AML), the most common form of leukemia amongst adults, is one of the most important hematological malignancies. Epidemiological data show both high incidence rates and low survival rates, especially in secondary cases among adults. Although classic and novel chemotherapeutic approaches have extensively improved disease prognosis and survival, the need for more personalized and target-specific methods with less side effects have been inevitable. Therefore, immunotherapeutic methods are of importance. In the following review, primarily a brief understanding of the molecular basis of the disease has been represented. Second, prior to the introduction of immunotherapeutic approaches, the entangled relationship of AML and patient's immune system has been discussed. At last, mechanistic and clinical evidence of each of the immunotherapy approaches have been covered.
Collapse
|
49
|
Lee WHS, Ye Z, Cheung AMS, Goh YPS, Oh HLJ, Rajarethinam R, Yeo SP, Soh MK, Chan EHL, Tan LK, Tan SY, Chuah C, Chng WJ, Connolly JE, Wang CI. Effective Killing of Acute Myeloid Leukemia by TIM-3 Targeted Chimeric Antigen Receptor T Cells. Mol Cancer Ther 2021; 20:1702-1712. [PMID: 34158344 DOI: 10.1158/1535-7163.mct-20-0155] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/11/2020] [Accepted: 06/01/2021] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive disease with poor outcomes, overwhelmingly due to relapse. Minimal residual disease (MRD), defined as the persistence of leukemic cells after chemotherapy treatment, is thought to be the major cause of relapse. The origins of relapse in AML have been traced to rare therapy-resistant leukemic stem cells (LSCs) that are already present at diagnosis. Effective treatment strategies for long-term remission are lacking, as it has been difficult to eliminate LSCs with conventional therapy. Here, we proposed a new approach based on the chimeric antigen receptor (CAR)-directed T lymphocytes, targeting T-cell immunoglobulin, and mucin domain 3 (TIM-3) to treat MRD in patients with AML. TIM-3 is selected as the target because it is highly expressed on AML blasts and LSCs in most subtypes regardless of the patient's genetic characteristics and treatment course. Moreover, it is absent in the normal hematopoietic stem cells, granulocytes, naïve lymphocytes, and most normal nonhematopoietic tissues. Using a naïve human Fab phage display library, we isolated an anti-human TIM-3 antibody and designed a second-generation anti-TIM-3. Our anti-TIM-3 CAR T cells exhibit potent antileukemic activity against AML cell lines and primary AML blasts, and in the mouse models. More importantly, we demonstrate efficient killing of the primary LSCs directly isolated from the patients. Hence, eradication of the LSCs present in the MRD by anti-TIM-3 CAR T-cell therapy following the first-line treatment may improve the clinical outcomes of patients with AML.
Collapse
Affiliation(s)
- Wen-Hsin Sandy Lee
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Zhiyong Ye
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Alice M S Cheung
- Department of Haematology, Singapore General Hospital, Singapore
| | - Y P Sharon Goh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Hsueh Ling Janice Oh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Ravisankar Rajarethinam
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Siok Ping Yeo
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Mun Kuen Soh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Esther Hian Li Chan
- Department of Haematology-Oncology, National University Health Systems, Singapore
| | - Lip Kun Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.,Department of Laboratory Medicine, National University Hospital, Singapore
| | - Soo-Yong Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, Singapore
| | - Charles Chuah
- Department of Haematology, Singapore General Hospital, Singapore.,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Wee Joo Chng
- Department of Haematology-Oncology, National University Health Systems, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - John E Connolly
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Institute of Biomedical Studies, Baylor University, Waco
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore.
| |
Collapse
|
50
|
CD19-specific CAR-T cell therapy for relapsed/refractory non-B-cell acute leukaemia with CD19 antigen expression. Eur J Cancer 2021; 153:1-4. [PMID: 34126332 DOI: 10.1016/j.ejca.2021.04.042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/21/2021] [Accepted: 04/30/2021] [Indexed: 11/21/2022]
|