1
|
Haubner S, Subklewe M, Sadelain M. Honing CAR T cells to tackle acute myeloid leukemia. Blood 2025; 145:1113-1125. [PMID: 39630061 DOI: 10.1182/blood.2024024063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/15/2024] [Indexed: 03/14/2025] Open
Abstract
ABSTRACT Acute myeloid leukemia (AML) remains a dismal disease with poor prognosis, particularly in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR) therapy has yielded remarkable clinical results in other leukemias and thus has, in principle, the potential to achieve similar outcomes in R/R AML. Redirecting the approved CD19-specific CAR designs against the myeloid antigens CD33, CD123, or CLEC12A has occasionally yielded morphologic leukemia-free states but has so far been marred by threatening myeloablation and early relapses. These safety and efficacy limitations are largely due to the challenge of identifying suitable target antigens and designing adequate receptors for effective recognition and safe elimination of AML. Building on lessons learned from the initial clinical attempts, a new wave of CAR strategies relying on alternative target antigens and innovative CAR designs is about to enter clinical evaluation. Adapted multiantigen targeting, logic gating, and emerging cell engineering solutions offer new possibilities to better direct T-cell specificity and sensitivity toward AML. Pharmacologic modulation and genetic epitope engineering may extend these approaches by augmenting target expression in AML cells or minimizing target expression in normal hematopoietic cells. On/off switches or CAR T-cell depletion may curb excessive or deleterious CAR activity. Investigation of AML-intrinsic resistance and leukemic microenvironmental factors is poised to reveal additional targetable AML vulnerabilities. We summarize here the findings, challenges, and new developments of CAR therapy for AML. These illustrate the need to specifically adapt CAR strategies to the complex biology of AML to achieve better therapeutic outcomes.
Collapse
Affiliation(s)
- Sascha Haubner
- Columbia Initiative in Cell Engineering and Therapy, Department of Medicine, Columbia University, New York, NY
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Michel Sadelain
- Columbia Initiative in Cell Engineering and Therapy, Department of Medicine, Columbia University, New York, NY
| |
Collapse
|
2
|
Soleimani Samarkhazan H, Zehtabcheh S, Seraji HR, Beqaj SH, Tayefeh S, Mohammadi MH, Aghaei M. Unveiling the potential of CLL-1: a promising target for AML therapy. Biomark Res 2025; 13:28. [PMID: 39940055 PMCID: PMC11823018 DOI: 10.1186/s40364-025-00738-6] [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: 10/08/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Acute myeloid leukemia (AML) remains a formidable blood cancer, despite recent advances in treatment. A significant challenge persists in improving patient outcomes, particularly in addressing relapse and treatment resistance. Identifying new therapeutic targets is critical for advancing AML therapy. C-type lectin-like molecule-1 (CLL-1) has emerged as a promising therapeutic target in AML. This cell surface receptor is highly expressed on AML blasts and demonstrates stable expression throughout disease progression. CLL-1's consistent presence makes it an ideal candidate for monitoring minimal residual disease (MRD), which is a critical indicator for predicting relapse. Beyond its utility as a diagnostic marker, CLL-1 offers exciting potential in the development of immunotherapies. Emerging strategies, such as CAR-T-cell therapy and antibody-drug conjugates (ADCs), are being investigated to leverage the immune system against CLL-1-expressing AML cells. This review examines the structure, function, and expression patterns of CLL-1 in AML and other hematologic malignancies, providing insights into its role in disease pathogenesis and treatment potential. Exploring CLL-1 as a target for diagnosis, MRD monitoring, and immunotherapy opens new avenues for AML treatment. A deeper understanding of its relationship with AML pathogenesis will aid in the development of targeted therapies, offering hope for improved patient outcomes in the future.
Collapse
Affiliation(s)
- Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Zehtabcheh
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamideh Rahmani Seraji
- Department of Hematology and Oncology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shamim Tayefeh
- UCLA Immunogenetics Center, Immunogenetics (UIC), 1000 Veteran Ave, Los Angeles, CA, 90024, USA
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
3
|
Zhu A, Bai Y, Nan Y, Ju D. Natural killer cell engagers: From bi-specific to tri-specific and tetra-specific engagers for enhanced cancer immunotherapy. Clin Transl Med 2024; 14:e70046. [PMID: 39472273 PMCID: PMC11521791 DOI: 10.1002/ctm2.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/25/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Natural killer cell engagers (NKCEs) are a specialised subset of antibodies capable of simultaneously targeting endogenous NK cells and tumour cells, generating precise and effective cytolytic responses against cancer. This review systematically explores NK engagers as a rising star in NK-mediated immunotherapy, specifically focusing on multi-specific engagers. It examines the diverse configuration of NKCEs and how certain biologics could be employed to boost NK activity, including activating receptor engagement and cytokine incorporation. Some challenges and future perspectives of current NKCEs therapy are also discussed, including optimising pharmacokinetics, addressing the immunosuppressive tumour microenvironment and exploring potential combinatorial approaches. By offering an in-depth analysis of the current landscape and future trajectories of multi-specific NKCEs in cancer treatment, this review serves as a valuable resource for understanding this promising field of immunotherapy. HIGHLIGHTS Innovative NKCEs: NK cell engagers (NKCEs) represent a promising new class of immunotherapeutics targeting tumours by activating NK cells. Multi-specific formats: The transition from bi-specific to multi-specific NKCEs enhances their versatility and therapeutic efficacy. MECHANISMS OF ACTION NKCEs have the potential to improve NK cell activation by engaging activating receptors and incorporating cytokines. CLINICAL POTENTIAL Current clinical trials demonstrate the safety and efficacy of various NKCEs across different cancer types. Future research directions: Optimising NKCE designs and exploring combination therapies are essential for overcoming challenges in cancer treatment.
Collapse
Affiliation(s)
- An Zhu
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Yu Bai
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of ImmunotherapeuticsFudan University School of PharmacyShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| |
Collapse
|
4
|
Kandav G, Chandel A. Revolutionizing cancer treatment: an in-depth exploration of CAR-T cell therapies. Med Oncol 2024; 41:275. [PMID: 39400611 DOI: 10.1007/s12032-024-02491-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/27/2024] [Indexed: 10/15/2024]
Abstract
Cancer is a leading cause of fatality worldwide. Due to the heterogeneity of cancer cells the effectiveness of various conventional cancer treatment techniques is constrained. Thus, researchers are diligently investigating therapeutic approaches like immunotherapy for effective tumor managements. Immunotherapy harnesses the inherent potential of patient's immune system to achieve desired outcomes. Within the realm of immunotherapy, CAR-T (Chimeric Antigen Receptor T) cells, emerges as a revolutionary innovation for cancer therapy. The process of CAR-T cell therapy entails extracting the patient's T cells, altering them with customized receptors designed to specifically recognize and eradicate the tumor cells, and then reinfusing the altered cells into the patient's body. Although there has been significant progress with CAR-T cell therapy in certain cases of specific B-cell leukemia and lymphoma, its effectiveness is hindered in hematological and solid tumors due to the challenges such as severe toxicities, restricted tumor infiltration, cytokine release syndrome and antigen escape. Overcoming these obstacles requires innovative approaches to design more effective CAR-T cells, which require a competent and diverse team to develop and implement. This comprehensive review addresses numerous therapeutic issues and provides a strategic solution while providing a deep understanding of the structural intricacies and production processes of CAR-T cells. In addition, this review explores the practical aspects of CAR-T cell therapy in clinical settings.
Collapse
Affiliation(s)
- Gurpreet Kandav
- Chandigarh College of Pharmacy, Chandigarh Group of Colleges, Landran, Sahibzada Ajit Singh Nagar, Punjab, 140307, India.
| | - Akash Chandel
- Chandigarh College of Pharmacy, Chandigarh Group of Colleges, Landran, Sahibzada Ajit Singh Nagar, Punjab, 140307, India
| |
Collapse
|
5
|
Mosna F. The Immunotherapy of Acute Myeloid Leukemia: A Clinical Point of View. Cancers (Basel) 2024; 16:2359. [PMID: 39001421 PMCID: PMC11240611 DOI: 10.3390/cancers16132359] [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: 06/02/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The potential of the immune system to eradicate leukemic cells has been consistently demonstrated by the Graft vs. Leukemia effect occurring after allo-HSCT and in the context of donor leukocyte infusions. Various immunotherapeutic approaches, ranging from the use of antibodies, antibody-drug conjugates, bispecific T-cell engagers, chimeric antigen receptor (CAR) T-cells, and therapeutic infusions of NK cells, are thus currently being tested with promising, yet conflicting, results. This review will concentrate on various types of immunotherapies in preclinical and clinical development, from the point of view of a clinical hematologist. The most promising therapies for clinical translation are the use of bispecific T-cell engagers and CAR-T cells aimed at lineage-restricted antigens, where overall responses (ORR) ranging from 20 to 40% can be achieved in a small series of heavily pretreated patients affected by refractory or relapsing leukemia. Toxicity consists mainly in the occurrence of cytokine-release syndrome, which is mostly manageable with step-up dosing, the early use of cytokine-blocking agents and corticosteroids, and myelosuppression. Various cytokine-enhanced natural killer products are also being tested, mainly as allogeneic off-the-shelf therapies, with a good tolerability profile and promising results (ORR: 20-37.5% in small trials). The in vivo activation of T lymphocytes and NK cells via the inhibition of their immune checkpoints also yielded interesting, yet limited, results (ORR: 33-59%) but with an increased risk of severe Graft vs. Host disease in transplanted patients. Therefore, there are still several hurdles to overcome before the widespread clinical use of these novel compounds.
Collapse
Affiliation(s)
- Federico Mosna
- Hematology and Bone Marrow Transplantation Unit (BMTU), Hospital of Bolzano (SABES-ASDAA), Teaching Hospital of Paracelsus Medical University (PMU), 39100 Bolzano, Italy
| |
Collapse
|
6
|
Zhong F, Yao F, Jiang J, Yu X, Liu J, Huang B, Wang X. CD8 + T cell-based molecular subtypes with heterogeneous immune landscapes and clinical significance in acute myeloid leukemia. Inflamm Res 2024; 73:329-344. [PMID: 38195768 DOI: 10.1007/s00011-023-01839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy. Although high-dose chemotherapy is the primary treatment option, it cannot cure the disease, and new approaches need to be developed. The tumor microenvironment (TME) plays a crucial role in tumor biology and immunotherapy. CD8 + T cells are the main anti-tumor immune effector cells, and it is essential to understand their relationship with the TME and the clinicopathological characteristics of AML. METHODS In this study, we conducted a systematic analysis of CD8 + T cell infiltration through multi-omics data and identified molecular subtypes with significant differences in CD8 + T cell infiltration and prognosis. We aimed to provide a comprehensive evaluation of the pathological factors affecting the prognosis of AML patients and to offer theoretical support for the precise treatment of AML. RESULTS Our results indicate that CD8 + T cell infiltration is accompanied by immunosuppression, and that there are two molecular subtypes, with the C2 subtype having a significantly worse prognosis than the C1 subtype, as well as less CD8 + T cell infiltration. We developed a signature to distinguish molecular subtypes using multiple machine learning algorithms and validated the prognostic predictive power of molecular subtypes in nine AML cohorts including 2059 AML patients. Our findings suggest that there are different immunosuppressive characteristics between the two subtypes. The C1 subtype has up-regulated expression of immune checkpoints such as CTLA4, PD-1, LAG3, and TIGITD, while the C2 subtype infiltrates more immunosuppressive cells such as Tregs and M2 macrophages. The C1 subtype is more responsive to anti-PD-1 immunotherapy and induction chemotherapy, as well as having higher immune and cancer-promoting variant-related pathway activity. Patients with the C2 subtype had a higher FLT3 mutation rate, higher WBC counts, and a higher percentage of blasts, as indicated by increased activity of signaling pathways involved in energy metabolism and cell proliferation. Analysis of data from ex vivo AML cell drug assays has identified a group of drugs that differ in therapeutic sensitivity between molecular subtypes. CONCLUSIONS Our results suggest that the molecular subtypes we constructed have potential application value in the prognosis evaluation and treatment guidance of AML patients.
Collapse
Affiliation(s)
- Fangmin Zhong
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Junyao Jiang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiajing Yu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jing Liu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
7
|
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
|
8
|
Guarnera L, Bravo-Perez C, Visconte V. Immunotherapy in Acute Myeloid Leukemia: A Literature Review of Emerging Strategies. Bioengineering (Basel) 2023; 10:1228. [PMID: 37892958 PMCID: PMC10604866 DOI: 10.3390/bioengineering10101228] [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: 09/15/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
In the last twenty years, we have witnessed a paradigm shift in the treatment and prognosis of acute myeloid leukemia (AML), thanks to the introduction of new efficient drugs or approaches to refine old therapies, such as Gemtuzumab Ozogamicin, CPX 3-5-1, hypomethylating agents, and Venetoclax, the optimization of conditioning regimens in allogeneic hematopoietic stem cell transplantation and the improvement of supportive care. However, the long-term survival of non-M3 and non-core binding factor-AML is still dismal. For this reason, the expectations for the recently developed immunotherapies, such as antibody-based therapy, checkpoint inhibitors, and chimeric antigen receptor strategies, successfully tested in other hematologic malignancies, were very high. The inherent characteristics of AML blasts hampered the development of these treatments, and the path of immunotherapy in AML has been bumpy. Herein, we provide a detailed review of potential antigenic targets, available data from pre-clinical and clinical trials, and future directions of immunotherapies in AML.
Collapse
Affiliation(s)
- Luca Guarnera
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlos Bravo-Perez
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER—Instituto de Salud Carlos III, 30005 Murcia, Spain
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
| |
Collapse
|
9
|
Lee E, Lee S, Park S, Son YG, Yoo J, Koh Y, Shin DY, Lim Y, Won J. Asymmetric anti-CLL-1×CD3 bispecific antibody, ABL602 2+1, with attenuated CD3 affinity endows potent antitumor activity but limited cytokine release. J Immunother Cancer 2023; 11:e007494. [PMID: 37848261 PMCID: PMC10582864 DOI: 10.1136/jitc-2023-007494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a type of leukemia in adults with a high mortality rate and poor prognosis. Although targeted therapeutics, chemotherapy, and hematopoietic stem cell transplantation can improve the prognosis, the recurrence rate is still high, with a 5-year survival rate of approximately 40%. This study aimed to develop an IgG-based asymmetric bispecific antibody that targets CLL-1 and CD3 for treating AML. METHODS ABL602 candidates were compared in terms of binding activity, T-cell activation, and tumor-killing activities. ABL602-mediated T-cell activation and tumor-killing activities were determined by measuring the expression of activation markers, cytokines, cytolytic proteins, and the proportion of dead cells. We evaluated in vivo tumor growth inhibitory activity in two mouse models bearing subcutaneously and orthotopically engrafted human AML. Direct tumor-killing activity and T-cell activation in patient-derived AML blasts were also evaluated. RESULTS ABL602 2+1 showed a limited CD3 binding in the absence of CLL-1, suggesting that steric hindrance on the CD3 binding arm could reduce CLL-1 expression-independent CD3 binding. Although the CD3 binding activity was attenuated compared with that of 1+1, ABL602 2+1 exhibited much stronger T-cell activation and potent tumor-killing activities in AML cell lines. ABL602 2+1 efficiently inhibited tumor progression in subcutaneously and orthotopically engrafted AML mouse models. In the orthotopic mouse model, tumor growth inhibition was observed by gross measurement of luciferase activity, as well as a reduced proportion of AML blasts in the bone marrow, as determined by flow cytometry and immunohistochemistry (IHC) staining. ABL602 2+1 efficiently activated T cells and induced the lysis of AML blasts, even at very low effector:target (E:T) ratios (eg, 1:50). Compared with the reference 1+1 antibody, ABL602 did not induce the release of cytokines including interleukin-6 and tumor necrosis factor-α in the healthy donor-derived peripheral blood mononuclear cell. CONCLUSIONS With its potent tumor-killing activity and reduced cytokine release, ABL602 2+1 is a promising candidate for treating patients with AML and warrants further study.
Collapse
Affiliation(s)
- Eunhee Lee
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Shinai Lee
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | | | | | - Jiseon Yoo
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Korea (the Republic of)
| | - Dong-Yeop Shin
- Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Korea (the Republic of)
| | - Yangmi Lim
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| | - Jonghwa Won
- ABL Bio Inc, Seongnam, Korea (the Republic of)
| |
Collapse
|
10
|
Xie D, Jin X, Sun R, Zhang M, Lu W, Cao X, Guo R, Zhang Y, Zhao M. Bicistronic CAR-T cells targeting CD123 and CLL1 for AML to reduce the risk of antigen escape. Transl Oncol 2023; 34:101695. [PMID: 37224766 DOI: 10.1016/j.tranon.2023.101695] [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: 02/14/2023] [Revised: 04/15/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
PURPOSE Acute myeloid leukemia (AML) is a highly heterogeneous neoplastic disease with a poor prognosis that relapses even after its treatment with chimeric antigen receptor (CAR)-T cells targeting a single antigen. CD123 and CLL1 are expressed in most AML blasts and leukemia stem cells, and their low expression in normal hematopoietic stem cells makes them ideal targets for CAR-T. In this study, we tested the hypothesis that a new bicistronic CAR targeting CD123 and CLL1 can enhance antigenic coverage and prevent antigen escape and subsequent recurrence of AML. METHODS CD123 and CLL1 expressions were evaluated on AML cell lines and blasts. Then, in addition to concentrating on CD123 and CLL1, we introduced the marker/suicide gene RQR8 with a bicistronic CAR. Xenograft models of disseminated AML and in vitro coculture models were used to assess the anti-leukemia efficacy of CAR-T cells. The hematopoietic toxicity of CAR-T cells was evaluated in vitro by colony cell formation assays. It was demonstrated in vitro that the combination of rituximab and NK cells caused RQR8-mediated clearance of 123CL CAR-T cells. RESULTS We have successfully established bicistronic 123CL CAR-T cells that can target CD123 and CLL1. 123CL CAR-T cells effectively cleared AML cell lines and blasts. They also demonstrated appreciable anti-AML activity in animal transplant models. Moreover, 123CL CAR-T cells can be eliminated in an emergency by a natural safety switch and don't target hematopoietic stem cells. CONCLUSIONS The bicistronic CAR-T cells targeting CD123 and CLL1 may be a useful and secure method for treating AML.
Collapse
Affiliation(s)
- Danni Xie
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Xin Jin
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Rui Sun
- Nankai University School of Medicine, Tianjin, China
| | - Meng Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, No. 24, Fukang Road, Nankai District, Tianjin 300190, China
| | - Xinping Cao
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Ruiting Guo
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Yi Zhang
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Mingfeng Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin, China; Nankai University School of Medicine, Tianjin, China; Department of Hematology, Tianjin First Central Hospital, No. 24, Fukang Road, Nankai District, Tianjin 300190, China.
| |
Collapse
|
11
|
Abbasi S, Totmaj MA, Abbasi M, Hajazimian S, Goleij P, Behroozi J, Shademan B, Isazadeh A, Baradaran B. Chimeric antigen receptor T (CAR-T) cells: Novel cell therapy for hematological malignancies. Cancer Med 2023; 12:7844-7858. [PMID: 36583504 PMCID: PMC10134288 DOI: 10.1002/cam4.5551] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/23/2022] [Accepted: 12/03/2022] [Indexed: 12/31/2022] Open
Abstract
Over the last decade, the emergence of several novel therapeutic approaches has changed the therapeutic perspective of human malignancies. Adoptive immunotherapy through chimeric antigen receptor T cell (CAR-T), which includes the engineering of T cells to recognize tumor-specific membrane antigens and, as a result, death of cancer cells, has created various clinical benefits for the treatment of several human malignancies. In particular, CAR-T-cell-based immunotherapy is known as a critical approach for the treatment of patients with hematological malignancies such as acute lymphoblastic leukemia (ALL), multiple myeloma (MM), chronic lymphocytic leukemia (CLL), acute myeloid leukemia (AML), Hodgkin lymphoma (HL), and non-Hodgkin's lymphoma (NHL). However, CAR-T-cell therapy of hematological malignancies is associated with various side effects. There are still extensive challenges in association with further progress of this therapeutic approach, from manufacturing and engineering issues to limitations of applications and serious toxicities. Therefore, further studies are required to enhance efficacy and minimize adverse events. In the current review, we summarize the development of CAR-T-cell-based immunotherapy and current clinical antitumor applications to treat hematological malignancies. Furthermore, we will mention the current advantages, disadvantages, challenges, and therapeutic limitations of CAR-T-cell therapy.
Collapse
Affiliation(s)
- Samane Abbasi
- Department of Biology, Faculty of SciencesUniversity of GuilanRashtIran
| | - Milad Asghari Totmaj
- Department of Clinical Immunology, Faculty of MedicineThe University of ManchesterManchesterUK
| | - Masoumeh Abbasi
- Department of Microbiology, Malekan BranchIslamic Azad UniversityMalekanIran
| | - Saba Hajazimian
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Pouya Goleij
- Department of Genetics, Faculty of BiologySana Institute of Higher EducationSariIran
| | - Javad Behroozi
- Department of Genetics and Biotechnology, School of MedicineAJA University of Medical SciencesTehranIran
| | - Behrouz Shademan
- Department of Medical Biology, Faculty of MedicineEge UniversityIzmirTurkey
| | - Alireza Isazadeh
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
12
|
Ciotti G, Marconi G, Sperotto A, Giannini MB, Gottardi M, Martinelli G. Biological therapy in elderly patients with acute myeloid leukemia. Expert Opin Biol Ther 2023; 23:175-194. [PMID: 36715330 DOI: 10.1080/14712598.2023.2174015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION The introduction of target molecules and immunological therapies is changing the treatment landscape of acute myeloid leukemia (AML). AREAS COVERED We recapitulate the biological therapies that can be employed in the treatment of elderly patients with AML. Alongside small molecules inhibitors that target specific gene mutations, antibodies, tumor microenvironment modulators, and cellular therapies are being developed for the cure of the disease. Here, we report the biological activities, the efficacy and toxicities of humanized antibodies and antibody-drug conjugates that targets surface antigens as CD33 (gemtuzumab ozogamicine) or CD123 (pivekimab sunirine). We further explore mechanisms and effectiveness of medications that modify the microenvironment, such as glasdegib, or that harness the immune system against leukemia, such as CD47 antibody magrolimab, PD1/PDL1 inhibitors pembrolizumab and nivolumab, TIM3 inhibitor sabatolimab, T-cell and NK-cell engagers. Cellular therapies are considered, even if a large trial is still pending for the feasibility of the approach. In this scenario, a brief overview of the mechanism of action of target agents is provided, particularly with respect to their biological mechanisms. EXPERT OPINION Overall, this therapeutic armamentarium will constitute the basis for multimodal and personalized combinations that, in the idea of precision medicine, will enormously benefit elderly AML patients.
Collapse
Affiliation(s)
- Giulia Ciotti
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Marconi
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alessandra Sperotto
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Maria B Giannini
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Gottardi
- Onco Hematology, Department of Oncology, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo Per Lo Studio Dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
13
|
Schorr C, Perna F. Targets for chimeric antigen receptor T-cell therapy of acute myeloid leukemia. Front Immunol 2022; 13:1085978. [PMID: 36605213 PMCID: PMC9809466 DOI: 10.3389/fimmu.2022.1085978] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is an aggressive myeloid malignancy associated with high mortality rates (less than 30% 5-year survival). Despite advances in our understanding of the molecular mechanisms underpinning leukemogenesis, standard-of-care therapeutic approaches have not changed over the last couple of decades. Chimeric Antigen Receptor (CAR) T-cell therapy targeting CD19 has shown remarkable clinical outcomes for patients with acute lymphoblastic leukemia (ALL) and is now an FDA-approved therapy. Targeting of myeloid malignancies that are CD19-negative with this promising technology remains challenging largely due to lack of alternate target antigens, complex clonal heterogeneity, and the increased recognition of an immunosuppressive bone marrow. We carefully reviewed a comprehensive list of AML targets currently being used in both proof-of-concept pre-clinical and experimental clinical settings. We analyzed the expression profile of these molecules in leukemic as well normal tissues using reliable protein databases and data reported in the literature and we provide an updated overview of the current clinical trials with CAR T-cells in AML. Our study represents a state-of-art review of the field and serves as a potential guide for selecting known AML-associated targets for adoptive cellular therapies.
Collapse
Affiliation(s)
- Christopher Schorr
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States,Department of Biomedical Engineering, Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, United States
| | - Fabiana Perna
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States,*Correspondence: Fabiana Perna,
| |
Collapse
|
14
|
Li Q, Liang C, Xu X, Zhang C, Cao W, Wang M, Jiang Z, Xing H, Yu J. CLEC12A plays an important role in immunomodulatory function and prognostic significance of patients with acute myeloid leukemia. Leuk Lymphoma 2022; 63:2136-2148. [PMID: 35481814 DOI: 10.1080/10428194.2022.2064986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/21/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
The physiological function and prognostic significance of C-type lectin domain family 12 member A (CLEC12A) in acute myeloid leukemia (AML) patients are unclear. CLEC12A transcriptional expression in a variety of tumors from several public databases was collected and compared. We found that CLEC12A was highly expressed in AML cell lines and in tissues from AML patients and a higher CLEC12A expression in leukemia stem cells. CLEC12A low expression was associated with poor prognosis in the chemotherapy-only group and high CLEC12A expression may benefit from autologous or allogeneic hematopoietic stem cell transplantation (HSCT). CLEC12A expression was positively correlated with infiltrating levels of type 2 macrophages and monocytes and negatively associated with NK cells and regulatory T cells in AML. CLEC12A high was positively associated with immune checkpoint genes as well as macrophage associated genes. CLEC12A is an ideal chimeric antigen receptor T-cell (CAR-T) therapy target for AML and its expression level was closely linked to treatment response and patients' survival outcome. CLEC12A plays an important immunomodulatory role in AML.
Collapse
MESH Headings
- Hematopoietic Stem Cell Transplantation
- Humans
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Prognosis
- Receptors, Chimeric Antigen
- Receptors, Mitogen/genetics
- Receptors, Mitogen/metabolism
Collapse
Affiliation(s)
- Qiaoqiao Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Chunyan Liang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xintong Xu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Congli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haizhou Xing
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, China
| |
Collapse
|
15
|
Jin X, Zhang M, Sun R, Lyu H, Xiao X, Zhang X, Li F, Xie D, Xiong X, Wang J, Lu W, Zhang H, Zhao M. First-in-human phase I study of CLL-1 CAR-T cells in adults with relapsed/refractory acute myeloid leukemia. J Hematol Oncol 2022; 15:88. [PMID: 35799191 PMCID: PMC9264641 DOI: 10.1186/s13045-022-01308-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/28/2022] [Indexed: 11/10/2022] Open
Abstract
Relapsed or refractory (R/R) acute myeloid leukemia (AML) has a poor prognosis. In this study, we evaluated chimeric antigen receptor (CAR) T cell therapy targeting CLL-1 in adults with R/R AML patients. Patients received conditioning chemotherapy with cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) for 3 days and an infusion of a dose of 1–2 × 106 CAR-T cells/kg. The incidence of dose-limiting toxicity was the primary endpoint. Ten patients were treated, and all developed cytokine release syndrome (CRS); 4 cases were low-grade, while the remaining 6 were considered high-grade CRS. No patient developed CAR-T cell-related encephalopathy syndrome (CRES). Severe pancytopenia occurred in all patients. Two patients died of severe infection due to chronic agranulocytosis. The complete response (CR)/CR with incomplete hematologic recovery (CRi) rate was 70% (n = 7/10). The median follow-up time was 173 days (15–488), and 6 patients were alive at the end of the last follow-up. CAR-T cells showed peak expansion within 2 weeks. Notably, CLL-1 is also highly expressed in normal granulocytes, so bridging hematopoietic stem cell transplantation (HSCT) may be a viable strategy to rescue long-term agranulocytosis due to off-target toxicity. In conclusion, this study is the first to demonstrate the positive efficacy and tolerable safety of CLL-1 CAR-T cell therapy in adult R/R AML.
Collapse
Affiliation(s)
- Xin Jin
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Meng Zhang
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Rui Sun
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China.,School of Medicine, Nankai University, Tianjin, 300071, China
| | - Hairong Lyu
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Xia Xiao
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Xiaomei Zhang
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China.,School of Medicine, Nankai University, Tianjin, 300071, China
| | - Fan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, 300350, China
| | - Danni Xie
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Xia Xiong
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Jiaxi Wang
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Wenyi Lu
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China.
| | - Hongkai Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Science, Nankai University, Tianjin, 300350, China.
| | - Mingfeng Zhao
- Tianjin First Central Hospital, The First Affiliated Central Hospital of Nankai University, The First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China. .,School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
16
|
Alnefaie A, Albogami S, Asiri Y, Ahmad T, Alotaibi SS, Al-Sanea MM, Althobaiti H. Chimeric Antigen Receptor T-Cells: An Overview of Concepts, Applications, Limitations, and Proposed Solutions. Front Bioeng Biotechnol 2022; 10:797440. [PMID: 35814023 PMCID: PMC9256991 DOI: 10.3389/fbioe.2022.797440] [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: 10/18/2021] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Adaptive immunity, orchestrated by B-cells and T-cells, plays a crucial role in protecting the body from pathogenic invaders and can be used as tools to enhance the body's defense mechanisms against cancer by genetically engineering these immune cells. Several strategies have been identified for cancer treatment and evaluated for their efficacy against other diseases such as autoimmune and infectious diseases. One of the most advanced technologies is chimeric antigen receptor (CAR) T-cell therapy, a pioneering therapy in the oncology field. Successful clinical trials have resulted in the approval of six CAR-T cell products by the Food and Drug Administration for the treatment of hematological malignancies. However, there have been various obstacles that limit the use of CAR T-cell therapy as the first line of defense mechanism against cancer. Various innovative CAR-T cell therapeutic designs have been evaluated in preclinical and clinical trial settings and have demonstrated much potential for development. Such trials testing the suitability of CARs against solid tumors and HIV are showing promising results. In addition, new solutions have been proposed to overcome the limitations of this therapy. This review provides an overview of the current knowledge regarding this novel technology, including CAR T-cell structure, different applications, limitations, and proposed solutions.
Collapse
Affiliation(s)
- Alaa Alnefaie
- Department of Medical Services, King Faisal Medical Complex, Taif, Saudi Arabia
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Yousif Asiri
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Saqer S. Alotaibi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Mohammad M. Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Hisham Althobaiti
- Chief of Medical Department, King Faisal Medical Complex (KFMC), Taif, Saudi Arabia
| |
Collapse
|
17
|
Perna F, Espinoza-Gutarra MR, Bombaci G, Farag SS, Schwartz JE. Immune-Based Therapeutic Interventions for Acute Myeloid Leukemia. Cancer Treat Res 2022; 183:225-254. [PMID: 35551662 DOI: 10.1007/978-3-030-96376-7_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive, clonally heterogeneous, myeloid malignancy, with a 5-year overall survival of approximately 27%. It constitutes the most common acute leukemia in adults, with an incidence of 3-5 cases per 100,000 in the United States. Despite great advances in understanding the molecular mechanisms underpinning leukemogenesis, the past several decades had seen little change to the backbone of therapy, comprised of an anthracycline-based induction regimen for those who are fit enough to receive it, followed by risk-stratified post-remission therapy with consolidation cytarabine or allogeneic stem cell transplantation (allo-SCT). Allo-SCT is the most fundamental form of immunotherapy in which donor cytotoxic T and NK cells recognize and eradicate residual AML in the graft-versus-leukemia (GvL) effect. Building on that, several alternative or synergistic approaches to exploit both self and foreign immunity against AML have been developed. Checkpoint inhibitors, for example, CTLA-4 inhibitors, PD-1 inhibitors, and PD-L1 inhibitors block proteins found on T cells or cancer cells that stop the immune system from attacking the cancer cells. They have been used with limited success in both the AML relapsed/refractory (R/R) and post SCT settings. AML tumor mutational burden is low compared to solid tumors and thus, it is less likely to generate neoantigens and respond to antibody-mediated checkpoint blockade that has shown unprecedented results in solid tumors. Therefore, alternative therapeutic strategies that work independently of the T cell receptor (TCR) specificity have been developed. They include bispecific antibodies, which recruit T cells through CD3 engagement, and in AML have shown an overall response rate ranging between 14 and 30% in early phase trials. Chimeric Antigen Receptor (CAR) T cell therapy is a type of treatment in which T cells are genetically engineered to produce a recombinant receptor that redirects the specificity and function of T lymphocytes. However, lack of cell surface targets exclusively expressed on AML cells including Leukemic Stem Cells (LSCs) combined with clonal heterogeneity represents the biggest challenge in developing CAR therapy for AML. Antibody-Drug Conjugates (ADC) constitute the only FDA-approved immunotherapy to treat AML with Gemtuzumab Ozogamicin, a CD33-specific ADC used in CEBPα-mutated AML. The identification of additional cell surface targets is critical for the development of other ADC's potentially useful in the induction and maintenance regimens, given the ease at which these reagents can be generated and managed. Here, we will review those immune-based therapeutic interventions and highlight active areas of research investigations toward fulfillment of the great promise of immunotherapy to AML.
Collapse
Affiliation(s)
- Fabiana Perna
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA.
| | - Manuel R Espinoza-Gutarra
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| | - Giuseppe Bombaci
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| | - Sherif S Farag
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| | - Jennifer E Schwartz
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, USA
| |
Collapse
|
18
|
Combinatorial antigen targeting strategies for acute leukemia: application in myeloid malignancy. Cytotherapy 2022; 24:282-290. [PMID: 34955406 PMCID: PMC8950815 DOI: 10.1016/j.jcyt.2021.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/06/2021] [Accepted: 10/11/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND AIMS Efforts to safely and effectively treat acute myeloid leukemia (AML) by targeting a single leukemia-associated antigen with chimeric antigen receptor (CAR) T cells have met with limited success, due in part to heterogeneous expression of myeloid antigens. The authors hypothesized that T cells expressing CARs directed toward two different AML-associated antigens would eradicate tumors and prevent relapse. METHODS For co-transduction with the authors' previously optimized CLL-1 CAR currently in clinical study (NCT04219163), the authors generated two CARs targeting either CD123 or CD33. The authors then tested the anti-tumor activity of T cells expressing each of the three CARs either alone or after co-transduction. The authors analyzed CAR T-cell phenotype, expansion and transduction efficacy and assessed function by in vitro and in vivo activity against AML cell lines expressing high (MOLM-13: CD123 high, CD33 high, CLL-1 intermediate), intermediate (HL-60: CD123 low, CD33 intermediate, CLL-1 intermediate/high) or low (KG-1a: CD123 low, CD33 low, CLL-1 low) levels of the target antigens. RESULTS The in vitro benefit of dual expression was most evident when the target cell line expressed low antigen levels (KG-1a). Mechanistically, dual expression was associated with higher pCD3z levels in T cells compared with single CAR T cells on exposure to KG-1a (P < 0.0001). In vivo, combinatorial targeting with CD123 or CD33 and CLL-1 CAR T cells improved tumor control and animal survival for all lines (KG-1a, MOLM-13 and HL-60); no antigen escape was detected in residual tumors. CONCLUSIONS Overall, these findings demonstrate that combinatorial targeting of CD33 or CD123 and CLL-1 with CAR T cells can control growth of heterogeneous AML tumors.
Collapse
|
19
|
[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
|
20
|
The Fusion of CLEC12A and MIR223HG Arises from a trans-Splicing Event in Normal and Transformed Human Cells. Int J Mol Sci 2021; 22:ijms222212178. [PMID: 34830054 PMCID: PMC8625150 DOI: 10.3390/ijms222212178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 11/17/2022] Open
Abstract
Chimeric RNAs are often associated with chromosomal rearrangements in cancer. In addition, they are also widely detected in normal tissues, contributing to transcriptomic complexity. Despite their prevalence, little is known about the characteristics and functions of chimeric RNAs. Here, we examine the genetic structure and biological roles of CLEC12A-MIR223HG, a novel chimeric transcript produced by the fusion of the cell surface receptor CLEC12A and the miRNA-223 host gene (MIR223HG), first identified in chronic myeloid leukemia (CML) patients. Surprisingly, we observed that CLEC12A-MIR223HG is not just expressed in CML, but also in a variety of normal tissues and cell lines. CLEC12A-MIR223HG expression is elevated in pro-monocytic cells resistant to chemotherapy and during monocyte-to-macrophage differentiation. We observed that CLEC12A-MIR223HG is a product of trans-splicing rather than a chromosomal rearrangement and that transcriptional activation of CLEC12A with the CRISPR/Cas9 Synergistic Activation Mediator (SAM) system increases CLEC12A-MIR223HG expression. CLEC12A-MIR223HG translates into a chimeric protein, which largely resembles CLEC12A but harbours an altered C-type lectin domain altering key disulphide bonds. These alterations result in differences in post-translational modifications, cellular localization, and protein-protein interactions. Taken together, our observations support a possible involvement of CLEC12A-MIR223HG in the regulation of CLEC12A function. Our workflow also serves as a template to study other uncharacterized chimeric RNAs.
Collapse
|
21
|
Aitken MJL, Ravandi F, Patel KP, Short NJ. Prognostic and therapeutic implications of measurable residual disease in acute myeloid leukemia. J Hematol Oncol 2021; 14:137. [PMID: 34479626 PMCID: PMC8417965 DOI: 10.1186/s13045-021-01148-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/25/2021] [Indexed: 01/10/2023] Open
Abstract
Quantification of measurable residual disease (MRD) provides critical prognostic information in acute myeloid leukemia (AML). A variety of platforms exist for MRD detection, varying in their sensitivity and applicability to individual patients. MRD detected by quantitative polymerase chain reaction, multiparameter flow cytometry, or next-generation sequencing has prognostic implications in various subsets of AML and at various times throughout treatment. While it is overwhelmingly evident that minute levels of remnant disease confer increased risk of relapse and shortened survival, the therapeutic implications of MRD remain less clear. The use of MRD as a guide to selecting the most optimal post-remission therapy, including hematopoietic stem cell transplant or maintenance therapy with hypomethylating agents, small molecule inhibitors, or immunotherapy is an area of active investigation. In addition, whether there are sufficient data to use MRD negativity as a surrogate endpoint in clinical trial development is controversial. In this review, we will critically examine the methods used to detect MRD, its role as a prognostic biomarker, MRD-directed therapeutics, and its potential role as a study endpoint.
Collapse
Affiliation(s)
- Marisa J L Aitken
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,McGovern Medical School, UT Health Science Center-Houston, Houston, TX, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Bi-specific and Tri-specific NK Cell Engagers: The New Avenue of Targeted NK Cell Immunotherapy. Mol Diagn Ther 2021; 25:577-592. [PMID: 34327614 DOI: 10.1007/s40291-021-00550-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2021] [Indexed: 02/01/2023]
Abstract
Natural killer (NK) cell-mediated cancer immunotherapy has grown significantly over the past two decades. More recently, multi-specific engagers have been developed as cancer therapeutics to effectively arm endogenous NK cells to more potently induce specific cytolytic responses against tumor targets. This review explores the bi- and tri-specific NK/tumor engagers that are emerging as a new generation of immunotherapeutics. These molecules vary in configuration, but they typically have small molecular weights and domains that engage specific tumor antigens and NK cell-activating receptors such as CD16, NKp30, NKp46, and NKG2D. They have demonstrated compelling potential in boosting NK cell cytotoxicity against specific tumor targets. This highly adaptable off-the-shelf platform, which in some formats also integrates cytokines, is poised to revolutionize targeted NK cell immunotherapy, either as a monotherapy or in combination with other effective anti-cancer therapies.
Collapse
|
23
|
Masoumi J, Jafarzadeh A, Abdolalizadeh J, Khan H, Philippe J, Mirzaei H, Mirzaei HR. Cancer stem cell-targeted chimeric antigen receptor (CAR)-T cell therapy: Challenges and prospects. Acta Pharm Sin B 2021; 11:1721-1739. [PMID: 34386318 PMCID: PMC8343118 DOI: 10.1016/j.apsb.2020.12.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/03/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) with their self-renewal ability are accepted as cells which initiate tumors. CSCs are regarded as interesting targets for novel anticancer therapeutic agents because of their association with tumor recurrence and resistance to conventional therapies, including radiotherapy and chemotherapy. Chimeric antigen receptor (CAR)-T cells are engineered T cells which express an artificial receptor specific for tumor associated antigens (TAAs) by which they accurately target and kill cancer cells. In recent years, CAR-T cell therapy has shown more efficiency in cancer treatment, particularly regarding blood cancers. The expression of specific markers such as TAAs on CSCs in varied cancer types makes them as potent tools for CAR-T cell therapy. Here we review the CSC markers that have been previously targeted with CAR-T cells, as well as the CSC markers that may be used as possible targets for CAR-T cell therapy in the future. Furthermore, we will detail the most important obstacles against CAR-T cell therapy and suggest solutions.
Collapse
Affiliation(s)
- Javad Masoumi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan 77181759111, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Jeandet Philippe
- Research Unit “Induced Resistance and Plant Bioprotection”, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences University of Reims Champagne-Ardenne, BP 1039, 51687, Reims Cedex 2, France
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan 8713781147, Iran
- Corresponding authors. Tel./fax: +98 31 55540022; Tel./fax: +98 21 66419536.
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Corresponding authors. Tel./fax: +98 31 55540022; Tel./fax: +98 21 66419536.
| |
Collapse
|
24
|
Daver N, Alotaibi AS, Bücklein V, Subklewe M. T-cell-based immunotherapy of acute myeloid leukemia: current concepts and future developments. Leukemia 2021; 35:1843-1863. [PMID: 33953290 PMCID: PMC8257483 DOI: 10.1038/s41375-021-01253-x] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/09/2021] [Accepted: 04/06/2021] [Indexed: 02/01/2023]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease linked to a broad spectrum of molecular alterations, and as such, long-term disease control requires multiple therapeutic approaches. Driven largely by an improved understanding and targeting of these molecular aberrations, AML treatment has rapidly evolved over the last 3-5 years. The stellar successes of immunotherapies that harness the power of T cells to treat solid tumors and an improved understanding of the immune systems of patients with hematologic malignancies have led to major efforts to develop immunotherapies for the treatment of patients with AML. Several immunotherapies that harness T cells against AML are in various stages of preclinical and clinical development. These include bispecific and dual antigen receptor-targeting antibodies (targeted to CD33, CD123, CLL-1, and others), chimeric antigen receptor (CAR) T-cell therapies, and T-cell immune checkpoint inhibitors (including those targeting PD-1, PD-L1, CTLA-4, and newer targets such as TIM3 and STING). The current and future directions of these T-cell-based immunotherapies in the treatment landscape of AML are discussed in this review.
Collapse
Affiliation(s)
- Naval Daver
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA.
| | - Ahmad S Alotaibi
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX, USA
- Oncology Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Veit Bücklein
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU Munich, Munich, Germany.
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
25
|
Ngai LL, Ma CY, Maguire O, Do AD, Robert A, Logan AC, Griffiths EA, Nemeth MJ, Green C, Pourmohamad T, van Kuijk BJ, Snel AN, Kwidama ZW, Venniker-Punt B, Cooper J, Manz MG, Gjertsen BT, Smit L, Ossenkoppele GJ, Janssen JJWM, Cloos J, Sumiyoshi T. Bimodal expression of potential drug target CLL-1 (CLEC12A) on CD34+ blasts of AML patients. Eur J Haematol 2021; 107:343-353. [PMID: 34053123 PMCID: PMC8457079 DOI: 10.1111/ejh.13672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/26/2021] [Indexed: 12/17/2022]
Abstract
Objectives This study aims to retrospectively assess C‐lectin‐like molecule 1 (CLL‐1) bimodal expression on CD34+ blasts in acute myeloid leukemia (AML) patients (total N = 306) and explore potential CLL‐1 bimodal associations with leukemia and patient‐specific characteristics. Methods Flow cytometry assays were performed to assess the deeper immunophenotyping of CLL‐1 bimodality. Cytogenetic analysis was performed to characterize the gene mutation on CLL‐1‐negative subpopulation of CLL‐1 bimodal AML samples. Results The frequency of a bimodal pattern of CLL‐1 expression of CD34+ blasts ranged from 8% to 65% in the different cohorts. Bimodal CLL‐1 expression was most prevalent in patients with MDS‐related AML (P = .011), ELN adverse risk (P = .002), NPM1 wild type (WT, P = .049), FLT3 WT (P = .035), and relatively low percentages of leukemia‐associated immunophenotypes (P = .006). Additional immunophenotyping analysis revealed the CLL‐1− subpopulation may consist of pre‐B cells, immature myeloblasts, and hematopoietic stem cells. Furthermore, (pre)‐leukemic mutations were detected in both CLL‐1+ and CLL‐1− subfractions of bimodal samples (N = 3). Conclusions C‐lectin‐like molecule 1 bimodality occurs in about 25% of AML patients and the CLL‐1− cell population still contains malignant cells, hence it may potentially limit the effectiveness of CLL‐1‐targeted therapies and warrant further investigation.
Collapse
Affiliation(s)
- Lok Lam Ngai
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Connie Y Ma
- Department of Development of Science, Genentech, South San Francisco, CA, USA
| | - Orla Maguire
- Flow and Image Cytometry Shared Resource, Buffalo, NY, USA
| | - An D Do
- Department of Development of Science, Genentech, South San Francisco, CA, USA
| | - Alberto Robert
- Department of Development of Science, Genentech, South San Francisco, CA, USA
| | - Aaron C Logan
- Department of Medicine, UCSF, San Francisco, CA, USA
| | | | | | - Cherie Green
- Department of Development of Science, Genentech, South San Francisco, CA, USA
| | | | - Bo J van Kuijk
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alexander N Snel
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Zinia W Kwidama
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Bianca Venniker-Punt
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - James Cooper
- Department of Early Clinical Development, Genentech, South San Francisco, CA, USA
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University and University Hospital, Zurich, Switzerland
| | - Bjørn T Gjertsen
- Section for Hematology, Institute of Clinical Science, University of Bergen, Bergen, Norway
| | - Linda Smit
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Gert J Ossenkoppele
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jeroen J W M Janssen
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jacqueline Cloos
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Teiko Sumiyoshi
- Department of Development of Science, Genentech, South San Francisco, CA, USA
| |
Collapse
|
26
|
Challenges and Solutions to Bringing Chimeric Antigen Receptor T-Cell Therapy to Myeloid Malignancies. ACTA ACUST UNITED AC 2021; 27:143-150. [PMID: 33750074 DOI: 10.1097/ppo.0000000000000512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Myeloid malignancies including myelodysplastic syndromes and acute myeloid leukemia are a group of clonal hematopoietic stem progenitor cell disorders mainly effecting the elderly. Chemotherapeutic approaches improved the outcome in majority of the patients, but it is generally associated with severe toxicities and relapse and does not benefit all the patients. With the success of adoptive cell therapies including chimeric antigen receptor T-cell therapy in treating certain B-cell malignancies, these therapeutic approaches are also being tested for myeloid malignancies, but the preclinical and limited clinical trial data suggest there are significant challenges. The principal hurdle to efficient targeted immunotherapy approaches is the lack of a unique targetable antigen on cancer cells leading to off-target effects including myelosuppression due to depletion of normal myeloid cells. Advanced age of the patients, comorbidities, immunosuppressive bone marrow microenvironment, and cytokine release syndrome are some other challenges that are not unique to myeloid malignancies but pose significant challenge for the successful adaptation of this approach for treatment. In this review, we highlight the challenges and solutions to adopt chimeric antigen receptor T-cell therapies to treat myeloid malignancies.
Collapse
|
27
|
A new era of immuno-oncology in acute myeloid leukemia - antibody-based therapies and immune checkpoint inhibition. Best Pract Res Clin Haematol 2020; 33:101220. [PMID: 33279176 DOI: 10.1016/j.beha.2020.101220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/28/2020] [Indexed: 01/02/2023]
Abstract
Acute myeloid leukemia (AML) remains a therapeutically challenging malignancy with high rate of relapse and poor outcomes. There has been increased understanding of the molecular characteristics of AML and the various roles of the immune system in its pathogenesis, the result of which has led to the study and development of multiple immune-based approaches for this disease. In this review, we aim to provide an overview of the recent advancements made in antibody-based approaches to the treatment of AML including monoclonal antibodies, antibody-drug conjugates, and immune checkpoint inhibition. In addition, we provide insight and discuss the promise of these agents, some of which may soon enter the therapeutic armamentarium we currently employ against this lethal disease.
Collapse
|
28
|
Acharya UH, Walter RB. Chimeric Antigen Receptor (CAR)-Modified Immune Effector Cell Therapy for Acute Myeloid Leukemia (AML). Cancers (Basel) 2020; 12:E3617. [PMID: 33287224 PMCID: PMC7761730 DOI: 10.3390/cancers12123617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Despite the availability of an increasing number of targeted therapeutics and wider use of allogeneic hematopoietic stem cell transplantation, many patients with acute myeloid leukemia (AML) ultimately succumb to this disease. Given their remarkable efficacy in B-acute lymphoblastic leukemia and other CD19-expressing B cell malignancies, there is hope adoptive cellular transfer, particularly chimeric antigen receptor (CAR)-modified immune effector cell (IEC) therapies, may afford a novel, potent immune-based approach for the treatment of AML that complements or replaces existing ones and improves cure rates. However, it is unclear how best to translate the success of these therapies from B cell malignancies, where use of highly potent immunotherapies is facilitated by identified target antigens with near ubiquitous expression on malignant cells and non-fatal consequences from "on-target, off-tumor cell" toxicities. Herein, we review the current status of CAR-modified IEC therapies for AML, with considerations regarding suitable, relatively leukemia-restricted target antigens, expected toxicities, and interactions of the engineered cells with a profoundly immunosuppressive tumor microenvironment that restricts their therapeutic efficacy. With these challenges in mind, we will discuss possible strategies to improve the cells' potency as well as their therapeutic window for optimal clinical use in AML.
Collapse
Affiliation(s)
- Utkarsh H. Acharya
- Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
29
|
Arvindam US, van Hauten PMM, Schirm D, Schaap N, Hobo W, Blazar BR, Vallera DA, Dolstra H, Felices M, Miller JS. A trispecific killer engager molecule against CLEC12A effectively induces NK-cell mediated killing of AML cells. Leukemia 2020; 35:1586-1596. [PMID: 33097838 PMCID: PMC8189652 DOI: 10.1038/s41375-020-01065-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/18/2020] [Accepted: 10/09/2020] [Indexed: 02/08/2023]
Abstract
The low five-year survival rate for patients with acute myeloid leukemia (AML), primarily caused due to disease relapse, emphasizes the need for better therapeutic strategies. Disease relapse is facilitated by leukemic stem cells (LSCs) that are resistant to standard chemotherapy and promote tumor growth. To target AML blasts and LSCs using Natural Killer (NK) cells, we have developed a trispecific killer engager (TriKE™) molecule containing a humanized anti-CD16 heavy chain camelid single domain antibody (sdAb) that activates NK cells, an IL-15 molecule that drives NK cell priming, expansion and survival, and a single-chain variable fragment (scFv) against human CLEC12A (CLEC12A TriKE). CLEC12A is a myeloid lineage antigen that is highly expressed by AML cells and LSCs, but not expressed by normal hematopoietic stem cells (HSCs), thus minimizing off-target toxicity. The CLEC12A TriKE induced robust NK cell specific proliferation, enhanced NK cell activation and killing of both AML cell lines and primary patient derived AML blasts in vitro while sparing healthy HSCs. Additionally, the CLEC12A TriKE was able to reduce tumor burden in pre-clinical mouse models. These findings highlight the clinical potential of the CLEC12A TriKE for the effective treatment of AML.
Collapse
Affiliation(s)
- Upasana Sunil Arvindam
- Division of Adult and Pediatric Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Paulien M M van Hauten
- Department of Laboratory Medicine-Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dawn Schirm
- Division of Adult and Pediatric Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Nicolaas Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine-Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bruce R Blazar
- Division of Adult and Pediatric Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Daniel A Vallera
- Department of Therapeutic Radiology-Radiation Oncology, University of Minnesota, Minneapolis, MN, USA
| | - Harry Dolstra
- Department of Laboratory Medicine-Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Felices
- Division of Adult and Pediatric Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S Miller
- Division of Adult and Pediatric Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
30
|
Lappalainen M, Hämäläinen S, Romppanen T, Pulkki K, Pyörälä M, Koivula I, Jantunen E, Juutilainen A. Febrile neutropenia in patients with acute myeloid leukemia: Outcome in relation to qSOFA score, C-reactive protein, and blood culture findings. Eur J Haematol 2020; 105:731-740. [PMID: 32740997 DOI: 10.1111/ejh.13500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To evaluate quick Sequential Organ Failure Assessment (qSOFA) score during febrile neutropenia (FN) in adult patients receiving intensive chemotherapy for acute myeloid leukemia (AML). METHODS qSOFA score, as well as the association of qSOFA score with ICU admission, infectious mortality, blood culture findings, and C-reactive protein (CRP) measurements during FN were assessed among 125 adult AML patients with 355 FN periods receiving intensive chemotherapy in a tertiary care hospital from November 2006 to December 2018. RESULTS The multivariate model for qSOFA score ≥ 2 included CRP ≥ 150 mg/L on d0-2 [OR 2.9 (95% CI 1.1-7.3), P = .026], Gram-negative bacteremia [OR 2.7 (95% CI 1.1-6.9), P = .034], and treatment according to AML-2003 vs more recent protocols [OR 2.7 (95% CI 1.0-7.4), P = .047]. Age or gender did not gain significance in the model. qSOFA score ≥ 2 was associated with ICU treatment and infectious mortality during FN with sensitivity and specificity of 0.700 and 0.979, and 1.000 and 0.971, respectively. CONCLUSION qSOFA offers a useful tool to evaluate the risk of serious complications in AML patients during FN.
Collapse
Affiliation(s)
- Marika Lappalainen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine/Internal Medicine, Faculty of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sari Hämäläinen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Tuomo Romppanen
- Institute of Clinical Medicine/Internal Medicine, Faculty of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Kari Pulkki
- Laboratory Division, Turku University Hospital, Clinical Chemistry, Faculty of Medicine, University of Turku, Turku, Finland.,Eastern Finland Laboratory Centre, Kuopio, Finland
| | - Marja Pyörälä
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Irma Koivula
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Esa Jantunen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine/Internal Medicine, Faculty of Medicine, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, North Carelia Central Hospital, Joensuu, Finland
| | - Auni Juutilainen
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland.,Institute of Clinical Medicine/Internal Medicine, Faculty of Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
31
|
Ataca Atilla P, McKenna MK, Tashiro H, Srinivasan M, Mo F, Watanabe N, Simons BW, McLean Stevens A, Redell MS, Heslop HE, Mamonkin M, Brenner MK, Atilla E. Modulating TNFα activity allows transgenic IL15-Expressing CLL-1 CAR T cells to safely eliminate acute myeloid leukemia. J Immunother Cancer 2020; 8:jitc-2020-001229. [PMID: 32938629 PMCID: PMC7497527 DOI: 10.1136/jitc-2020-001229] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
Background C-type lectin-like molecule 1 (CLL-1) is highly expressed in acute myeloid leukemia (AML) but is absent in primitive hematopoietic progenitors, making it an attractive target for a chimeric antigen receptor (CAR) T-cell therapy. Here, we optimized our CLL-1 CAR for anti-leukemic activity in mouse xenograft models of aggressive AML. Methods First, we optimized the CLL-1 CAR using different spacer, transmembrane and costimulatory sequences. We used a second retroviral vector to coexpress transgenic IL15. We measured the effects of each construct on T cell phenotype and sequential (recursive) co culture assays with tumor cell targets to determine the durability of the anti tumor activity by flow cytometry. We administered CAR T cells to mice engrafted with patient derived xenografts (PDX) and AML cell line and determined anti tumor activity by bioluminescence imaging and weekly bleeding, measured serum cytokines by multiplex analysis. After euthanasia, we examined formalin-fixed/paraffin embedded sections. Unpaired two-tailed Student’s t-tests were used and values of p<0.05 were considered significant. Survival was calculated using Mantel-Cox log-rank test. Results In vitro, CLL-1 CAR T cells with interleukin-15 (IL15) were less terminally differentiated (p<0.0001) and had superior expansion compared with CD28z-CD8 CAR T cells without IL15 (p<0.001). In both AML PDX and AML cell line animal models, CLL-1 CAR T coexpressing transgenic IL15 initially expanded better than CD28z-CD8 CAR T without IL15 (p<0.0001), but produced severe acute toxicity associated with high level production of human tumor necrosis factor α (TNFα), IL15 and IL2. Histopathology showed marked inflammatory changes with tissue damage in lung and liver. This acute toxicity could be managed by two strategies, individually or in combination. The excessive TNF alpha secretion could be blocked with anti-TNF alpha antibody, while excessive T cell expansion could be arrested by activation of an inducible caspase nine safety switch by administration of dimerizing drug. Both strategies successfully prolonged tumor-free survival. Conclusion Combinatorial treatment with a TNFα blocking antibody and subsequent activation of the caspase-9 control switch increased the expansion, survival and antileukemic potency of CLL-1 CAR T-cells expressing transgenic IL15 while avoiding the toxicities associated with excessive cytokine production and long-term accumulation of activated T-cells.
Collapse
Affiliation(s)
- Pinar Ataca Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K McKenna
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Haruko Tashiro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | | | - Feiyan Mo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Norihiro Watanabe
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| | - Brian Wesley Simons
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Alexandra McLean Stevens
- Division of Pediatric Hematology/Oncology, Texas Children's Hospital, Houston, Texas, USA.,Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Michele S Redell
- Division of Pediatric Hematology/Oncology, Texas Children's Hospital, Houston, Texas, USA.,Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, Texas, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA.,Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Erden Atilla
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
32
|
Bewersdorf JP, Shallis RM, Boddu PC, Wood B, Radich J, Halene S, Zeidan AM. The minimal that kills: Why defining and targeting measurable residual disease is the “Sine Qua Non” for further progress in management of acute myeloid leukemia. Blood Rev 2020; 43:100650. [DOI: 10.1016/j.blre.2019.100650] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
|
33
|
Drouin M, Saenz J, Chiffoleau E. C-Type Lectin-Like Receptors: Head or Tail in Cell Death Immunity. Front Immunol 2020; 11:251. [PMID: 32133013 PMCID: PMC7040094 DOI: 10.3389/fimmu.2020.00251] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
C-type lectin-like receptors (CLRs) represent a family of transmembrane pattern recognition receptors, expressed primarily by myeloid cells. They recognize not only pathogen moieties for host defense, but also modified self-antigens such as damage-associated molecular patterns released from dead cells. Upon ligation, CLR signaling leads to the production of inflammatory mediators to shape amplitude, duration and outcome of the immune response. Thus, following excessive injury, dysregulation of these receptors leads to the development of inflammatory diseases. Herein, we will focus on four CLRs of the "Dectin family," shown to decode the immunogenicity of cell death. CLEC9A on dendritic cells links F-actin exposed by dying cells to favor cross-presentation of dead-cell associated antigens to CD8+ T cells. Nevertheless, CLEC9A exerts also feedback mechanisms to temper neutrophil recruitment and prevent additional tissue damage. MINCLE expressed by macrophages binds nuclear SAP130 released by necrotic cells to potentiate pro-inflammatory responses. However, the consequent inflammation can exacerbate pathogenesis of inflammatory diseases. Moreover, in a tumor microenvironment, MINCLE induces macrophage-induced immune suppression and cancer progression. Similarly, triggering of LOX-1 by oxidized LDL, amplifies pro-inflammatory response but promotes tumor immune escape and metastasis. Finally, CLEC12A that recognizes monosodium urate crystals formed during cell death, inhibits activating signals to prevent detrimental inflammation. Interestingly, CLEC12A also sustains type-I IFN response to finely tune immune responses in case of viral-induced collateral damage. Therefore, CLRs acting in concert as sensors of injury, could be used in a targeted way to treat numerous diseases such as allergies, obesity, tumors, and autoimmunity.
Collapse
Affiliation(s)
- Marion Drouin
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Javier Saenz
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Elise Chiffoleau
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
34
|
Mitchell K, Steidl U. Targeting Immunophenotypic Markers on Leukemic Stem Cells: How Lessons from Current Approaches and Advances in the Leukemia Stem Cell (LSC) Model Can Inform Better Strategies for Treating Acute Myeloid Leukemia (AML). Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036251. [PMID: 31451539 DOI: 10.1101/cshperspect.a036251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapies targeting cell-surface antigens in acute myeloid leukemia (AML) have been tested over the past 20 years with limited improvement in overall survival. Recent advances in the understanding of AML pathogenesis support therapeutic targeting of leukemia stem cells as the most promising avenue toward a cure. In this review, we provide an overview of the evolving leukemia stem cell (LSC) model, including evidence of the cell of origin, cellular and molecular disease architecture, and source of relapse in AML. In addition, we explore limitations of current targeted strategies utilized in AML and describe the various immunophenotypic antigens that have been proposed as LSC-directed therapeutic targets. We draw lessons from current approaches as well as from the (pre)-LSC model to suggest criteria that immunophenotypic targets should meet for more specific and effective elimination of disease-initiating clones, highlighting in detail a few targets that we suggest fit these criteria most completely.
Collapse
Affiliation(s)
- Kelly Mitchell
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Medicine (Oncology), Division of Hemato-Oncology, Albert Einstein College of Medicine-Montefiore Medical Center, Bronx, New York 10461, USA.,Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
35
|
Genome-scale CRISPR activation screen uncovers tumor-intrinsic modulators of CD3 bispecific antibody efficacy. Sci Rep 2019; 9:20068. [PMID: 31882897 PMCID: PMC6934601 DOI: 10.1038/s41598-019-56670-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
Bispecific antibodies (bsAb) that bridge tumor cells and CD3-positive effector T cells are being developed against many tumor cell targets. While tumor cell factors other than target expression level appear to play a role in determining the efficacy of CD3 bsAb, the identity of such factors remains largely unknown. Using a co-culture system of primary human T cells and B lymphoma cell lines, we demonstrate a range of sensitivities to CD20xCD3 bsAb that is independent of CD20 surface expression. To identify genes that modulate tumor cell sensitivity to CD3 bsAb, we employed a genome-scale CRISPR activation screen in a CD20xCD3-sensitive human B lymphoma cell line. Among the most highly enriched sgRNAs were those targeting genes with predicted effects on cell-cell adhesion, including sialophorin (SPN). Increased expression of SPN impeded tumor cell clustering with T cells, thereby limiting CD3 bsAb-mediated tumor cell lysis. This inhibitory effect of SPN appeared to be dependent on sialylated core 2 O-glycosylation of the protein. While SPN is not endogenously expressed in the majority of B cell lymphomas, it is highly expressed in acute myeloid leukemia. CRISPR-mediated SPN knockout in AML cell lines facilitated T cell-tumor cell clustering and enhanced CD3 bsAb-mediated AML cell lysis. In sum, our data establish that the cell cross-linking mechanism of CD3 bsAb is susceptible to subversion by anti-adhesive molecules expressed on the tumor cell surface. Further evaluation of anti-adhesive pathways may provide novel biomarkers of clinical response and enable the development of effective combination regimens for this promising therapeutic class.
Collapse
|
36
|
Yang X, Wang GX, Zhou JF. CAR T Cell Therapy for Hematological Malignancies. Curr Med Sci 2019; 39:874-882. [PMID: 31845217 DOI: 10.1007/s11596-019-2118-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/25/2019] [Indexed: 12/11/2022]
Abstract
As a rapidly progressing field in oncology, the adoptive transfer of T cells that have been genetically modified with chimeric antigen receptors (CARs) has shown striking efficacy in the management of hematological malignancies and has been reported in a number of clinical trials. Of note, CAR T cell therapy has shown extraordinary potential, especially in relapsed/refractory patients. However, there are still challenges regarding the further development of this strategy, spanning from engineering and manufacturing issues, to limited applications, to accompanying toxicities. In this review, we will summarize the general knowledge of this novel method, including receptor composition, applications, adverse events and challenges. Additionally, we will propose several comprehensive recommendations.
Collapse
Affiliation(s)
- Xin Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gao-Xiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Feng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
37
|
Tone K, Stappers MHT, Willment JA, Brown GD. C-type lectin receptors of the Dectin-1 cluster: Physiological roles and involvement in disease. Eur J Immunol 2019; 49:2127-2133. [PMID: 31580478 PMCID: PMC6916577 DOI: 10.1002/eji.201847536] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/12/2019] [Accepted: 09/27/2019] [Indexed: 12/27/2022]
Abstract
C-type lectin receptors (CLRs) are essential for multicellular existence, having diverse functions ranging from embryonic development to immune function. One subgroup of CLRs is the Dectin-1 cluster, comprising of seven receptors including MICL, CLEC-2, CLEC-12B, CLEC-9A, MelLec, Dectin-1, and LOX-1. Reflecting the larger CLR family, the Dectin-1 cluster of receptors has a broad range of ligands and functions, but importantly, is involved in numerous pathophysiological processes that regulate health and disease. Indeed, these receptors have been implicated in development, infection, regulation of inflammation, allergy, transplantation tolerance, cancer, cardiovascular disease, arthritis, and other autoimmune diseases. In this mini-review, we discuss the latest advancements in elucidating the function(s) of each of the Dectin-1 cluster CLRs, focussing on their physiological roles and involvement in disease.
Collapse
Affiliation(s)
- Kazuya Tone
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Aberdeen, Scotland
| | - Mark H T Stappers
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, Devon, England
| | - Janet A Willment
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Aberdeen, Scotland.,Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, Devon, England
| | - Gordon D Brown
- Aberdeen Fungal Group, University of Aberdeen, Institute of Medical Sciences, Aberdeen, Scotland.,Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, Devon, England
| |
Collapse
|
38
|
Chopra M, Bohlander SK. The cell of origin and the leukemia stem cell in acute myeloid leukemia. Genes Chromosomes Cancer 2019; 58:850-858. [PMID: 31471945 DOI: 10.1002/gcc.22805] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 12/19/2022] Open
Abstract
There is experimental and observational evidence that the cells of the leukemic clone in acute myeloid leukemia (AML) have different phenotypes even though they share the same somatic mutations. The organization of the malignant clone in AML has many similarities to normal hematopoiesis, with leukemia stem cells (LSCs) that sustain leukemia and give rise to more differentiated cells. LSCs, similar to normal hematopoietic stem cells (HSCs), are those cells that are able to give rise to a new leukemic clone when transplanted into a recipient. The cell of origin of leukemia (COL) is defined as the normal cell that is able to transform into a leukemia cell. Current evidence suggests that the COL is distinct from the LSC. Here, we will review the current knowledge about LSCs and the COL in AML.
Collapse
Affiliation(s)
- Martin Chopra
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Stefan K Bohlander
- Leukaemia & Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
39
|
Schinnerl D, Mejstrikova E, Schumich A, Zaliova M, Fortschegger K, Nebral K, Attarbaschi A, Fiser K, Kauer MO, Popitsch N, Haslinger S, Inthal A, Buldini B, Basso G, Bourquin JP, Gaipa G, Brüggemann M, Feuerstein T, Maurer-Granofszky M, Panzer-Grümayer R, Trka J, Mann G, Haas OA, Hrusak O, Dworzak MN, Strehl S. CD371 cell surface expression: a unique feature of DUX4-rearranged acute lymphoblastic leukemia. Haematologica 2019; 104:e352-e355. [PMID: 30705095 PMCID: PMC6669149 DOI: 10.3324/haematol.2018.214353] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Dagmar Schinnerl
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Ester Mejstrikova
- CLIP - Childhood Leukemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Angela Schumich
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Marketa Zaliova
- CLIP - Childhood Leukemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Klaus Fortschegger
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Karin Nebral
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital Medical University of Vienna, Vienna, Austria
| | - Karel Fiser
- CLIP - Childhood Leukemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Maximilian O Kauer
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Niko Popitsch
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Sabrina Haslinger
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Andrea Inthal
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Barbara Buldini
- Department of Woman's and Child's Health, Laboratory of Pediatric Oncohematology, University of Padova, Padova, Italy
| | - Giuseppe Basso
- Department of Woman's and Child's Health, Laboratory of Pediatric Oncohematology, University of Padova, Padova, Italy
| | - Jean-Pierre Bourquin
- Department of Oncology and Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - Giuseppe Gaipa
- Department of Pediatrics, University of Milano-Bicocca, Fondazione Tettamanti - Centro Ricerca M. Tettamanti, Monza, Italy
| | - Monika Brüggemann
- Department of Hematology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Tamar Feuerstein
- Pediatric Hematology Oncology, Schneider Children's Medical Center, Petah-Tikva, Israel
| | | | - Renate Panzer-Grümayer
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Jan Trka
- CLIP - Childhood Leukemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Georg Mann
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital Medical University of Vienna, Vienna, Austria
| | - Oskar A Haas
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| | - Ondrej Hrusak
- CLIP - Childhood Leukemia Investigation Prague, Prague, Czech Republic
- Department of Pediatric Hematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
- University Hospital Motol, Prague, Czech Republic
| | - Michael N Dworzak
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital Medical University of Vienna, Vienna, Austria
| | - Sabine Strehl
- CCRI, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Vienna, Austria
| |
Collapse
|
40
|
Morsink LM, Walter RB. Novel monoclonal antibody-based therapies for acute myeloid leukemia. Best Pract Res Clin Haematol 2019; 32:116-126. [PMID: 31203993 DOI: 10.1016/j.beha.2019.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/07/2019] [Indexed: 12/21/2022]
Abstract
There has been long-standing interest in using monoclonal antibodies to improve outcomes of people with acute myeloid leukemia (AML). While several candidate therapeutics have failed at various stages of clinical testing, improved survival of some patients receiving the CD33 antibody-drug conjugate gemtuzumab ozogamicin has provided first evidence that monoclonal antibodies have a role in the armamentarium against AML. Over the last several years, work to improve the success of monoclonal antibody-based therapies in AML has focused on the identification and exploration of new antigen targets as much as on the development of novel treatment formats such as use of unconjugated engineered monoclonal antibodies and conjugated antibodies, delivering highly potent small molecule drugs or radionuclides to AML cells. Here, we will provide a brief overview of current efforts with such investigational monoclonal antibody-based therapeutics.
Collapse
Affiliation(s)
- Linde M Morsink
- Department of Hematology, University Medical Center Groningen, Groningen, the Netherlands
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, Division of Hematology, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|