1
|
Rahnama R, Kizerwetter M, Yang H, Christodoulou I, Guaraca C, Holl NJ, Choe J, Vorri SC, Zinsky M, Jones DG, Garcia Espinoza N, Kuo YH, Zahurak M, Varadhan R, Spangler JB, Bonifant CL. Single-chain variable fragment affinity tuning can optimize anti-AML CAR-NK cell functionality. J Immunother Cancer 2025; 13:e010763. [PMID: 39915004 PMCID: PMC11804205 DOI: 10.1136/jitc-2024-010763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Natural Killer (NK) cells have intrinsic anticancer activity that can be redirected toward acute myeloid leukemia (AML) with chimeric antigen receptor (CAR) engineering. Here, we study the functional consequences of CAR binding affinity and targeted epitope on CAR-NK cell activation, cytolytic synapse formation, and antitumor activity. METHODS We characterized NK-92 and primary NK cell populations expressing variant affinity AML-specific CARs containing single-chain variable fragments (scFvs, 26292 or 7G3) targeting two epitopes on CD123. 26292 affinity variants were discovered through directed evolution of an error-prone mutagenic library, while 7G3 affinity variants were previously reported. The resulting CAR-NK cell panel was studied with in vitro binding, activation, and cytotoxicity studies and in mouse xenograft models. RESULTS 26292 and 7G3 CARs of variable CD123 binding affinities were highly expressed in NK cells and conferred antigen-specific activation in vitro. High-resolution imaging demonstrated greater clustering of high-affinity 7G3 CAR-NK cells and consequent AML target cell death in a short-term time lapse. Low-affinity 7G3 CAR-NK cells exhibited enhanced antigen density discrimination with greater membrane-proximal signaling, cytokine production, and cytotoxicity. In longer-term assays, low-affinity 7G3 CAR-NK cells demonstrated more sustained killing of AML cells. In vivo testing highlighted greater expansion of low-affinity 7G3 CAR-NK cells in two xenograft models. CONCLUSIONS Expression of 26292 and 7G3 CARs with a range of CD123 binding affinities in NK cells leads to antigen-specific activation and cytotoxicity against AML. Affinity-based differences in functional activation and antitumor activity are dependent on time course and are scFv/epitope specific.
Collapse
MESH Headings
- Humans
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Mice
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Single-Chain Antibodies/pharmacology
- Single-Chain Antibodies/immunology
- Immunotherapy, Adoptive/methods
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
Collapse
Affiliation(s)
- Ruyan Rahnama
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Monika Kizerwetter
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Huilin Yang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ilias Christodoulou
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Christian Guaraca
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Natalie Jordan Holl
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Jun Choe
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Stamatia C Vorri
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Megan Zinsky
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Danielle G Jones
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Nikol Garcia Espinoza
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yun-Huai Kuo
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marianna Zahurak
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Ravi Varadhan
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Jamie B Spangler
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Molecular Microbiology & Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Challice L Bonifant
- Oncology, Johns Hopkins Medicine Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
2
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Jiang X. RNA modification in normal hematopoiesis and hematologic malignancies. MedComm (Beijing) 2024; 5:e787. [PMID: 39445003 PMCID: PMC11496571 DOI: 10.1002/mco2.787] [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: 08/12/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant RNA modification in eukaryotic cells. Previous studies have shown that m6A plays a critical role under both normal physiological and pathological conditions. Hematopoiesis and differentiation are highly regulated processes, and recent studies on m6A mRNA methylation have revealed how this modification controls cell fate in both normal and malignant hematopoietic states. However, despite these insights, a comprehensive understanding of its complex roles between normal hematopoietic development and malignant hematopoietic diseases remains elusive. This review first provides an overview of the components and biological functions of m6A modification regulators. Additionally, it highlights the origin, differentiation process, biological characteristics, and regulatory mechanisms of hematopoietic stem cells, as well as the features, immune properties, and self-renewal pathways of leukemia stem cells. Last, the article systematically reviews the latest research advancements on the roles and mechanisms of m6A regulatory factors in normal hematopoiesis and related malignant diseases. More importantly, this review explores how targeting m6A regulators and various signaling pathways could effectively intervene in the development of leukemia, providing new insights and potential therapeutic targets. Targeting m6A modification may hold promise for achieving more precise and effective leukemia treatments.
Collapse
Affiliation(s)
- Xi Chen
- Department of NeurosurgeryThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Yixiao Yuan
- Department of MedicineUF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
- Department of Medicine and Department of Biochemistry and Molecular BiologyUniversity of FloridaGainesvilleFloridaUSA
| | - Fan Zhou
- Department of NeurosurgeryThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Jun Pu
- Department of NeurosurgeryThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingYunnanChina
| | - Xiulin Jiang
- Department of MedicineUF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
- Department of Medicine and Department of Biochemistry and Molecular BiologyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
3
|
Gao C, Li X, Xu Y, Zhang T, Zhu H, Yao D. Recent advances in CAR-T cell therapy for acute myeloid leukaemia. J Cell Mol Med 2024; 28:e18369. [PMID: 38712978 PMCID: PMC11075639 DOI: 10.1111/jcmm.18369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/18/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a fatal and refractory haematologic cancer that primarily affects adults. It interferes with bone marrow cell proliferation. Patients have a 5 years survival rate of less than 30% despite the availability of several treatments, including chemotherapy, allogeneic haematopoietic stem cell transplantation (Allo-HSCT), and receptor antagonist drugs. Allo-HSCT is the mainstay of acute myeloid leukaemia treatment. Although it does work, there are severe side effects, such as graft-versus-host disease (GVHD). In recent years, chimeric antigen receptor (CAR)-T cell therapies have made significant progress in the treatment of cancer. These engineered T cells can locate and recognize tumour cells in vivo and release a large number of effectors through immune action to effectively kill tumour cells. CAR-T cells are among the most effective cancer treatments because of this property. CAR-T cells have demonstrated positive therapeutic results in the treatment of acute myeloid leukaemia, according to numerous clinical investigations. This review highlights recent progress in new targets for AML immunotherapy, and the limitations, and difficulties of CAR-T therapy for AML.
Collapse
Affiliation(s)
- Chi Gao
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Xin Li
- College of BiotechnologyTianjin University of Science and TechnologyTianjinChina
| | - Yao Xu
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Tongcun Zhang
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
- Institute of Biology and MedicineWuhan University of Science and TechnologyWuhanChina
| | - Haichuan Zhu
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Di Yao
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| |
Collapse
|
4
|
Hou Z, Ren Y, Zhang X, Huang D, Yan F, Sun W, Zhang W, Zhang Q, Fu X, Lang Z, Chu C, Zou B, Gao B, Jin B, Kang Z, Liu Q, Yan J. EP300-ZNF384 transactivates IL3RA to promote the progression of B-cell acute lymphoblastic leukemia. Cell Commun Signal 2024; 22:211. [PMID: 38566191 PMCID: PMC10986138 DOI: 10.1186/s12964-024-01596-9] [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: 12/12/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
The EP300-ZNF384 fusion gene is an oncogenic driver in B-cell acute lymphoblastic leukemia (B-ALL). In the present study, we demonstrated that EP300-ZNF384 substantially induces the transcription of IL3RA and the expression of IL3Rα (CD123) on B-ALL cell membranes. Interleukin 3 (IL-3) supplementation promotes the proliferation of EP300-ZNF348-positive B-ALL cells by activating STAT5. Conditional knockdown of IL3RA in EP300-ZF384-positive cells inhibited the proliferation in vitro, and induced a significant increase in overall survival of mice, which is attributed to impaired propagation ability of leukemia cells. Mechanistically, the EP300-ZNF384 fusion protein transactivates the promoter activity of IL3RA by binding to an A-rich sequence localized at -222/-234 of IL3RA. Furthermore, forced EP300-ZNF384 expression induces the expression of IL3Rα on cell membranes and the secretion of IL-3 in CD19-positive B precursor cells derived from healthy individuals. Doxorubicin displayed a selective killing of EP300-ZNF384-positive B-ALL cells in vitro and in vivo. Collectively, we identify IL3RA as a direct downstream target of EP300-ZNF384, suggesting CD123 is a potent biomarker for EP300-ZNF384-driven B-ALL. Targeting CD123 may be a novel therapeutic approach to EP300-ZNF384-positive patients, alternative or, more likely, complementary to standard chemotherapy regimen in clinical setting.
Collapse
Affiliation(s)
- Zhijie Hou
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China.
| | - Yifei Ren
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China
| | - Xuehong Zhang
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, 116044, China
| | - Dan Huang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China
| | - Fanzhi Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China
| | - Wentao Sun
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Wenjuan Zhang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Qingqing Zhang
- Department of Pathology, Dalian Medical University, Dalian, 116044, China
| | - Xihui Fu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Zhenghui Lang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Chenyang Chu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Boyang Zou
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Beibei Gao
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China
| | - Bilian Jin
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China
| | - Zhijie Kang
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
| | - Quentin Liu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116044, China.
| | - Jinsong Yan
- Department of Hematology, Liaoning Medical Center for Hematopoietic Stem Cell Transplantation, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Liaoning Key Laboratory of Hematopoietic Stem Cell Transplantation and Translational Medicine, Dalian Key Laboratory of hematology, Diamond Bay institute of hematology, Blood Stem Cell Transplantation Institute, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
- Department of Pediatric, Pediatric Oncology and Hematology Center, the Second Hospital of Dalian Medical University, Dalian, 116027, China.
| |
Collapse
|
5
|
Sinha S, Hembram KC, Chatterjee S. Targeting signaling pathways in cancer stem cells: A potential approach for developing novel anti-cancer therapeutics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 385:157-209. [PMID: 38663959 DOI: 10.1016/bs.ircmb.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Cancer stem cells (CSCs) have emerged as prime players in the intricate landscape of cancer development, progression, and resistance to traditional treatments. These unique cellular subpopulations own the remarkable capability of self-renewal and differentiation, giving rise to the diverse cellular makeup of tumors and fostering their recurrence following conventional therapies. In the quest for developing more effective cancer therapeutics, the focus has now shifted toward targeting the signaling pathways that govern CSCs behavior. This chapter underscores the significance of these signaling pathways in CSC biology and their potential as pivotal targets for the development of novel chemotherapy approaches. We delve into several key signaling pathways essential for maintaining the defining characteristics of CSCs, including the Wnt, Hedgehog, Notch, JAK-STAT, NF-κB pathways, among others, shedding light on their potential crosstalk. Furthermore, we highlight the latest advancements in CSC-targeted therapies, spanning from promising preclinical models to ongoing clinical trials. A comprehensive understanding of the intricate molecular aspects of CSC signaling pathways and their manipulation holds the prospective to revolutionize cancer treatment paradigms. This, in turn, could lead to more efficacious and personalized therapies with the ultimate goal of eradicating CSCs and enhancing overall patient outcomes. The exploration of CSC signaling pathways represents a key step towards a brighter future in the battle against cancer.
Collapse
Affiliation(s)
- Saptarshi Sinha
- National Institute of Biomedical Genomics, Kalyani, West Bengal, India
| | | | - Subhajit Chatterjee
- Laboratory of Cellular Oncology, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, United States.
| |
Collapse
|
6
|
Ravandi F, Bashey A, Foran J, Stock W, Mawad R, Short N, Yilmaz M, Kantarjian H, Odenike O, Patel A, Garcha R, Ainsworth WB, Clynes R, Kanodia J, Ding Y, Li H, Kye S, Mims A. Phase 1 study of vibecotamab identifies an optimized dose for treatment of relapsed/refractory acute myeloid leukemia. Blood Adv 2023; 7:6492-6505. [PMID: 37647601 PMCID: PMC10632668 DOI: 10.1182/bloodadvances.2023010956] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023] Open
Abstract
Acute myeloid leukemia (AML), an aggressive malignancy with unmet medical need, lacks immunotherapeutic options. CD123, the cellular receptor for interleukin-3, expressed in AML is an attractive target for tumor-specific therapy. Vibecotamab (XmAb14045), a humanized bispecific antibody, monovalently binds both CD3 and CD123 to recruit cytotoxic T cells to kill CD123+ tumor cells. This phase 1 study's primary objectives were safety and tolerability and identification of a maximum tolerated dose/recommended dose for use as monotherapy in patients with relapsed/refractory AML. Identification of a recommended phase 2 vibecotamab dose comprised 3 step-up doses (Week 1), which were noted to reduce cytokine response syndrome (CRS), followed by weekly dosing (1.7 μg/kg, Cohort -1D). In 16 of 120 patients, at least 1 treatment-emergent adverse event was classified as a dose-limiting toxicity. CRS, the most common adverse event (59.2%), managed with premedication, were mostly ≤grade 2. A secondary objective was assessment of efficacy in patients with CD123-expressing leukemias. A total of 10 of 111 (9.0%) efficacy-evaluable patients with AML achieved an overall response of morphologic leukemia-free state or better with an overall objective response rate (ORR) of 9.0%. Response was only observed in patients receiving a target dose of 0.75 μg/kg or higher (n = 87) in which the efficacy-evaluable ORR was 11.5%. Response was associated with lower baseline blast counts in blood and bone marrow (<25%) suggesting potential benefit. This trial was registered at www.clinicaltrials.gov as #NCT02730312.
Collapse
Affiliation(s)
- Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Asad Bashey
- Blood and Marrow Transplant Program at Northside Hospital, Atlanta, GA
| | - James Foran
- Department of Hematology, Mayo Clinic, Jacksonville, FL
| | - Wendy Stock
- Section of Hematology/Oncology, The University of Chicago Department of Medicine, Chicago, IL
| | | | - Nicholas Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, The University of Chicago Department of Medicine, Chicago, IL
| | - Anand Patel
- Section of Hematology/Oncology, The University of Chicago Department of Medicine, Chicago, IL
| | | | | | | | | | | | | | | | - Alice Mims
- Wexner Medical Center at The Ohio State University James Cancer Hospital, Columbus, OH
| |
Collapse
|
7
|
Wen J, Chen Y, Liao C, Ma X, Wang M, Li Q, Wang D, Li Y, Zhang X, Li L, Zhou H, Zou J, Liu L, Peng D. Engineered mesenchymal stem cell exosomes loaded with miR-34c-5p selectively promote eradication of acute myeloid leukemia stem cells. Cancer Lett 2023; 575:216407. [PMID: 37769796 DOI: 10.1016/j.canlet.2023.216407] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/16/2023] [Accepted: 09/19/2023] [Indexed: 10/02/2023]
Abstract
Most patients with acute myeloid leukemia (AML) relapse eventually because of the inability to effectively eliminate leukemia stem cells (LSCs), prompting the search of new therapies to eradicate LSCs. Our previous study demonstrated that miR-34c-5p promotes the clearance of LSCs in an AML mouse model, highlighting its potential as a therapeutic target for eradicating LSCs, but the effective delivery of miR-34c-5p to LSCs remains a great challenge. Here, we employed simultaneous two-step modifications to engineer mesenchymal stem cells (MSCs) and MSC-derived exosomes to create exosomes overexpressing the fused protein lysosome-associated membrane protein 2-interleukin 3 (Lamp2b-IL3) and hematopoietic cell E-selectin/L-selectin ligand (HCELL), and demonstrated that the engineered exosomes exhibited an enhanced ability for bone marrow homing and selective targeting of LSCs. Additionally, using a humanized AML mouse model, we confirmed that the engineered exosomes, loaded with miR-34c-5p, could selectively promote eradication of LSCs and impede the AML development in vivo. In summary, we successfully designed an effective delivery system and provided new insights into the development of novel therapies for delivering miRNA or other molecules to LSCs with greater cellular targeting specificity.
Collapse
Affiliation(s)
- Jin Wen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenxi Liao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Ma
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengyuan Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingnan Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolan Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Li
- Department of Pediatrics, Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Danyue Peng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
8
|
Morse JW, Rios M, Ye J, Rios A, Zhang CC, Daver NG, DiNardo CD, Zhang N, An Z. Antibody therapies for the treatment of acute myeloid leukemia: exploring current and emerging therapeutic targets. Expert Opin Investig Drugs 2023; 32:107-125. [PMID: 36762937 PMCID: PMC10031751 DOI: 10.1080/13543784.2023.2179482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is the most common and deadly type of leukemia affecting adults. It is typically managed with rounds of non-targeted chemotherapy followed by hematopoietic stem cell transplants, but this is only possible in patients who can tolerate these harsh treatments and many are elderly and frail. With the identification of novel tumor-specific cell surface receptors, there is great conviction that targeted antibody therapies will soon become available for these patients. AREAS COVERED In this review, we describe the current landscape of known target receptors for monospecific and bispecific antibody-based therapeutics for AML. Here, we characterize each of the receptors and targeted antibody-based therapeutics in development, illustrating the rational design behind each therapeutic compound. We then discuss the bispecific antibodies in development and how they improve immune surveillance of AML. For each therapeutic, we also summarize the available pre-clinical and clinical data, including data from discontinued trials. EXPERT OPINION One antibody-based therapeutic has already been approved for AML treatment, the CD33-targeting antibody-drug conjugate, gemtuzumab ozogamicin. Many more are currently in pre-clinical and clinical studies. These antibody-based therapeutics can perform tumor-specific, elaborate cytotoxic functions and there is growing confidence they will soon lead to personalized, safe AML treatment options that induce durable remissions.
Collapse
Affiliation(s)
- Joshua W Morse
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Margarita Rios
- Gorgas Memorial Institute of Health Studies, Panama City, Panama
| | - John Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Adan Rios
- Division of Oncology, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
9
|
Liao Y, Xiong S, Ur Rehman Z, He X, Peng H, Liu J, Sun S. The Research Advances of Aptamers in Hematologic Malignancies. Cancers (Basel) 2023; 15:300. [PMID: 36612296 PMCID: PMC9818631 DOI: 10.3390/cancers15010300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Currently, research for hematological malignancies is very intensive, with many breakthroughs. Among them, aptamer-based targeted therapies could be counted. Aptamer is a targeting tool with many unique advantages (easy synthesis, low toxicity, easy modification, low immunogenicity, nano size, long stability, etc.), therefore many experts screened corresponding aptamers in various hematological malignancies for diagnosis and treatment. In this review, we try to summarize and provide the recent progress of aptamer research in the diagnosis and treatment of hematologic malignancies. Until now, 29 aptamer studies were reported in hematologic malignancies, of which 12 aptamers were tested in vivo and the remaining 17 aptamers were only tested in vitro. In this case, 11 aptamers were combined with chemotherapeutic drugs for the treatment of hematologic malignancies, 4 aptamers were used in combination with nanomaterials for the diagnosis and treatment of hematologic malignancies, and some studies used aptamers for the targeted transportation of siRNA and miRNA for targeted therapeutic effects. Their research provides multiple approaches to achieve more targeted goals. These findings show promising and encouraging future for both hematological malignancies basic and clinical trials research.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shuming Sun
- Department of Hematology, The Second Xiangya Hospital, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha 410011, China
| |
Collapse
|
10
|
Zhuo X, Dai H, Yu S. The cGAS-STING pathway-related gene signature can predict patient prognosis and immunotherapy responses in prostate adenocarcinoma. Medicine (Baltimore) 2022; 101:e31290. [PMID: 36550819 PMCID: PMC9771290 DOI: 10.1097/md.0000000000031290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The cyclic GMP-AMP synthase-stimulator of the interferon genes (cGAS-STING) pathway is essential in inflammation-driven tumor occurrence and progression. However, the prognostic roles and immune functions of cGAS-STING pathway-related genes in patients with prostate adenocarcinoma (PRAD) remain unclear. cGAS-STING pathway-related genes were obtained from the gene set enrichment analysis (GSEA) website. Univariate Cox regression analysis was performed to screen the prognosis-related hub genes in the cancer genome atlas (TCGA) and GSE116918 datasets. Unsupervised clustering analysis was performed to identify different clusters. The least absolute shrinkage and selection operator and multivariate Cox regression analyses were applied to develop a prognostic risk model. The prognostic values and predictive performance of risk signature were assessed by the Kaplan-Meier curve and receiver operating characteristic curve. The IMvigor210 cohort was used to investigate the potential values of the risk score in immunotherapeutic responses. Two clusters were identified based on the expression matrix of 12 prognosis-related genes. Specifically, better overall survival was observed in cluster 2 than cluster 1 in both datasets. Inflammation-related pathway enrichment and immune cell infiltration levels were altered between 2 clusters. Moreover, 6 genes (CASP8, GRK6, IL3RA, PLCB1, TBKBP1, and TNFSF10) were identified to generate a cGAS-STING pathway-related signature (CPRS). Survival analysis showed that patients in the high-risk group showed a more dismal survival than those in the low-risk group in TCGA and GSE116918 datasets. Notably, the CPRS can differentiate responsive patients from non-responsive individuals treated with PD-L1 blockades in an independent cohort. In addition, higher CPRS was associated with a more favorable prognosis. The proposed risk model was developed based on 6 cGAS-STING pathway related-genes, which can be used as a promising predictor for patient survival and immunotherapeutic responses in PRAD, contributing to treatment strategy-related decision-making.
Collapse
Affiliation(s)
- Xingxing Zhuo
- Department of Urology, Fenghua District People’s Hospital, Ningbo, People’s Republic of China
| | - Hao Dai
- Department of Urology, Fenghua District People’s Hospital, Ningbo, People’s Republic of China
| | - Sui Yu
- Department of Urology, Fenghua District People’s Hospital, Ningbo, People’s Republic of China
- *Correspondence: Sui Yu, Department of Urology, Fenghua District People’s Hospital, Ningbo 310053, People’s Republic of China (e-mail: )
| |
Collapse
|
11
|
Bhattacharjee R, Ghosh S, Nath A, Basu A, Biswas O, Patil CR, Kundu CN. Theragnostic strategies harnessing the self-renewal pathways of stem-like cells in the acute myeloid leukemia. Crit Rev Oncol Hematol 2022; 177:103753. [PMID: 35803452 DOI: 10.1016/j.critrevonc.2022.103753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 06/21/2022] [Accepted: 07/02/2022] [Indexed: 02/07/2023] Open
Abstract
Acute myelogenous leukemia (AML) is a genetically heterogeneous and aggressive cancer of the Hematopoietic Stem/progenitor cells. It is distinguished by the uncontrollable clonal growth of malignant myeloid stem cells in the bone marrow, venous blood, and other body tissues. AML is the most predominant of leukemias occurring in adults (25%) and children (15-20%). The relapse after chemotherapy is a major concern in the treatment of AML. The overall 5-year survival rate in young AML patients is about 40-45% whereas in the elderly patients it is less than 10%. Leukemia stem-like cells (LSCs) having the ability to self-renew indefinitely, repopulate and persist longer in the G0/G1 phase play a crucial role in the AML relapse and refractoriness to chemotherapy. Hence, novel treatment strategies and diagnostic biomarkers targeting LSCs are being increasingly investigated. Through this review, we have explored the signaling modulations in the LSCs as the theragnostic targets. The significance of the self-renewal pathways in overcoming the treatment challenges in AML has been highlighted.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sharad Ghosh
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Arijit Nath
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Asmita Basu
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Ojaswi Biswas
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Chandragauda R Patil
- Department of Pharmacology, DIPSAR, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Chanakya Nath Kundu
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India.
| |
Collapse
|
12
|
Cancer Stem Cells: From an Insight into the Basics to Recent Advances and Therapeutic Targeting. Stem Cells Int 2022; 2022:9653244. [PMID: 35800881 PMCID: PMC9256444 DOI: 10.1155/2022/9653244] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 06/07/2022] [Indexed: 12/22/2022] Open
Abstract
Cancer is characterized by an abnormal growth of the cells in an uncontrolled manner. These cells have the potential to invade and can eventually turn into malignancy, leading to highly fatal forms of tumor. Small subpopulations of cancer cells that are long-lived with the potential of excessive self-renewal and tumor formation are called cancer stem cells (CSCs) or cancer-initiating cells or tumor stem cells. CSCs can be found in tissues, such as breast, brain, lung, liver, ovary, and testis; however, their origin is still a matter of debate. These cells can differentiate and possess self-renewal capacity maintained by numerous intracellular signal transduction pathways, such as the Wnt/β-catenin signaling, Notch signaling, transforming growth factor-β signaling, and Hedgehog signaling. They can also contribute to numerous malignancies and are an important reason for tumor recurrence and metastasis because they are resistant to the known therapeutic strategies that mainly target the bulk of the tumor cells. This review contains collected and compiled information after analyzing published works of the last three decades. The goal was to gather information of recent breakthroughs related to CSCs, strategies to target CSCs' niche (e.g., nanotechnology with tumor biology), and their signaling pathways for cancer therapy. Moreover, the role of metformin, an antidiabetic drug, acting as a chemotherapeutic agent on CSCs by inhibiting cellular transformation and its selective killing is also addressed.
Collapse
|
13
|
Andreozzi F, Massaro F, Wittnebel S, Spilleboudt C, Lewalle P, Salaroli A. New Perspectives in Treating Acute Myeloid Leukemia: Driving towards a Patient-Tailored Strategy. Int J Mol Sci 2022; 23:3887. [PMID: 35409248 PMCID: PMC8999556 DOI: 10.3390/ijms23073887] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022] Open
Abstract
For decades, intensive chemotherapy (IC) has been considered the best therapeutic option for treating acute myeloid leukemia (AML), with no curative option available for patients who are not eligible for IC or who have had failed IC. Over the last few years, several new drugs have enriched the therapeutic arsenal of AML treatment for both fit and unfit patients, raising new opportunities but also new challenges. These include the already approved venetoclax, the IDH1/2 inhibitors enasidenib and ivosidenib, gemtuzumab ozogamicin, the liposomal daunorubicin/cytarabine formulation CPX-351, and oral azacitidine. Venetoclax, an anti BCL2-inhibitor, in combination with hypomethylating agents (HMAs), has markedly improved the management of unfit and elderly patients from the perspective of improved quality of life and better survival. Venetoclax is currently under investigation in combination with other old and new drugs in early phase trials. Recently developed drugs with different mechanisms of action and new technologies that have already been investigated in other settings (BiTE and CAR-T cells) are currently being explored in AML, and ongoing trials should determine promising agents, more synergic combinations, and better treatment strategies. Access to new drugs and inclusion in clinical trials should be strongly encouraged to provide scientific evidence and to define the future standard of treatment in AML.
Collapse
Affiliation(s)
- Fabio Andreozzi
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Fulvio Massaro
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Sebastian Wittnebel
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Chloé Spilleboudt
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Philippe Lewalle
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| | - Adriano Salaroli
- Hematology Department, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Bruxelles, Belgium; (F.M.); (S.W.); (C.S.); (P.L.); (A.S.)
| |
Collapse
|
14
|
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
|
15
|
Emerging CAR T Cell Strategies for the Treatment of AML. Cancers (Basel) 2022; 14:cancers14051241. [PMID: 35267549 PMCID: PMC8909045 DOI: 10.3390/cancers14051241] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Chimeric antigen receptors (CARs) targeting CD19 have emerged as a new treatment for hematological malignancies. As a “living therapy”, CARs can precisely target and eliminate tumors while proliferating inside the patient’s body. Various preclinical and clinical studies are ongoing to identify potential CAR-T cell targets for acute myeloid leukemia (AML). We shed light on the continuing efforts of CAR development to overcome tumor escape, exhaustion, and toxicities. Furthermore, we summarize the recent progress of a range of putative targets exploring this unmet need to treat AML. Lastly, we discuss the advances in preclinical models that built the foundation for ongoing clinical trials. Abstract Engineered T cells expressing chimeric antigen receptors (CARs) on their cell surface can redirect antigen specificity. This ability makes CARs one of the most promising cancer therapeutic agents. CAR-T cells for treating patients with B cell hematological malignancies have shown impressive results. Clinical manifestation has yielded several trials, so far five CAR-T cell therapies have received US Food and Drug Administration (FDA) approval. However, emerging clinical data and recent findings have identified some immune-related toxicities due to CAR-T cell therapy. Given the outcome and utilization of the same proof of concept, further investigation in other hematological malignancies, such as leukemias, is warranted. This review discusses the previous findings from the pre-clinical and human experience with CAR-T cell therapy. Additionally, we describe recent developments of novel targets for adoptive immunotherapy. Here we present some of the early findings from the pre-clinical studies of CAR-T cell modification through advances in genetic engineering, gene editing, cellular programming, and formats of synthetic biology, along with the ongoing efforts to restore the function of exhausted CAR-T cells through epigenetic remodeling. We aim to shed light on the new targets focusing on acute myeloid leukemia (AML).
Collapse
|
16
|
Insights into Modern Therapeutic Approaches in Pediatric Acute Leukemias. Cells 2022; 11:cells11010139. [PMID: 35011701 PMCID: PMC8749975 DOI: 10.3390/cells11010139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 02/01/2023] Open
Abstract
Pediatric cancers predominantly constitute lymphomas and leukemias. Recently, our knowledge and awareness about genetic diversities, and their consequences in these diseases, have greatly expanded. Modern solutions are focused on mobilizing and impacting a patient’s immune system. Strategies to stimulate the immune system, to prime an antitumor response, are of intense interest. Amid those types of therapies are chimeric antigen receptor T (CAR-T) cells, bispecific antibodies, and antibody–drug conjugates (ADC), which have already been approved in the treatment of acute lymphoblastic leukemia (ALL)/acute myeloid leukemia (AML). In addition, immune checkpoint inhibitors (ICIs), the pattern recognition receptors (PRRs), i.e., NOD-like receptors (NLRs), Toll-like receptors (TLRs), and several kinds of therapy antibodies are well on their way to showing significant benefits for patients with these diseases. This review summarizes the current knowledge of modern methods used in selected pediatric malignancies and presents therapies that may hold promise for the future.
Collapse
|
17
|
Loss of MBD2 attenuates MLL-AF9-driven leukemogenesis by suppressing the leukemic cell cycle via CDKN1C. Oncogenesis 2021; 10:79. [PMID: 34789717 PMCID: PMC8599466 DOI: 10.1038/s41389-021-00366-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/19/2021] [Accepted: 10/25/2021] [Indexed: 11/09/2022] Open
Abstract
Acute myeloid leukemia (AML) is a deadly cancer characterized by an expanded self-renewal capacity that is associated with the accumulation of immature myeloid cells. Emerging evidence shows that methyl-CpG-binding domain protein 2 (MBD2), a DNA methylation reader, often participates in the transcriptional silencing of hypermethylated genes in cancer cells. Nevertheless, the role of MBD2 in AML remains unclear. Herein, by using an MLL-AF9 murine model and a human AML cell line, we observed that loss of MBD2 could delay the initiation and progression of leukemia. MBD2 depletion significantly reduced the leukemia burden by decreasing the proportion of leukemic stem cells (LSCs) and inhibiting leukemia cell proliferation in serial transplantation experiments, thereby allowing leukemic blasts to transition to a more mature state reflecting normal myelopoiesis. Both gene expression analyses and bioinformatic studies revealed that MBD2 negatively modulated genes related to myeloid differentiation, and was necessary to sustain the MLL-AF9 oncogene-induced gene program. We further demonstrated that MBD2 could promote LSC cell cycle progression through epigenetic regulation of CDKN1C transcription probably by binding to its promoter region. Taken together, our data suggest that MBD2 promotes AML development and could be a therapeutic target for myeloid malignancies.
Collapse
|
18
|
Abstract
The β common chain (βc) cytokine family includes granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and IL-5, all of which use βc as key signaling receptor subunit. GM-CSF, IL-3 and IL-5 have specific roles as hematopoietic growth factors. IL-3 binds with high affinity to the IL-3 receptor α (IL-3Rα/CD123) and then associates with the βc subunit. IL-3 is mainly synthesized by different subsets of T cells, but is also produced by several other immune [basophils, dendritic cells (DCs), mast cells, etc.] and non-immune cells (microglia and astrocytes). The IL-3Rα is also expressed by immune (basophils, eosinophils, mast cells, DCs, monocytes, and megacaryocytes) and non-immune cells (endothelial cells and neuronal cells). IL-3 is the most important growth and activating factor for human and mouse basophils, primary effector cells of allergic disorders. IL-3-activated basophils and mast cells are also involved in different chronic inflammatory disorders, infections, and several types of cancer. IL-3 induces the release of cytokines (i.e., IL-4, IL-13, CXCL8) from human basophils and preincubation of basophils with IL-3 potentiates the release of proinflammatory mediators and cytokines from IgE- and C5a-activated basophils. IL-3 synergistically potentiates IL-33-induced mediator release from human basophils. IL-3 plays a pathogenic role in several hematologic cancers and may contribute to autoimmune and cardiac disorders. Several IL-3Rα/CD123 targeting molecules have shown some efficacy in the treatment of hematologic malignancies.
Collapse
|
19
|
Bonnevaux H, Guerif S, Albrecht J, Jouannot E, De Gallier T, Beil C, Lange C, Leuschner WD, Schneider M, Lemoine C, Caron A, Amara C, Barrière C, Siavellis J, Bardet V, Luna E, Agrawal P, Drake DR, Rao E, Wonerow P, Carrez C, Blanc V, Hsu K, Wiederschain D, Fraenkel PG, Virone-Oddos A. Pre-clinical development of a novel CD3-CD123 bispecific T-cell engager using cross-over dual-variable domain (CODV) format for acute myeloid leukemia (AML) treatment. Oncoimmunology 2021; 10:1945803. [PMID: 34484869 PMCID: PMC8409758 DOI: 10.1080/2162402x.2021.1945803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Novel therapies are needed for effective treatment of AML. In the relapsed setting, prognosis is very poor despite salvage treatment with chemotherapy. Evidence suggests that leukemic stem cells (LSCs) cause relapse. The cell surface receptor CD123 is highly expressed in blast cells and LSCs from AML patients and is a potential therapeutic target. CD123 cross-over dual-variable domain T-cell engager (CD123-CODV-TCE) is a bispecific antibody with an innovative format. One arm targets the CD3εδ subunit of T-cell co-receptors on the surface of T cells, while the other targets CD123 on malignant cells, leading to cell-specific cytotoxic activity. Here, we describe the preclinical activity of CD123-CODV-TCE. CD123-CODV-TCE effectively binds to human and cynomolgus monkey CD3 and CD123 and is a highly potent T-cell engager. It mediates T-cell activation and T-cell-directed killing of AML cells in vitro. In vivo, CD123-CODV-TCE suppresses AML tumor growth in leukemia xenograft mouse models, where it achieves an effective half-life of 3.2 days, which is a significantly longer half-life compared to other bispecific antibodies with no associated Fc fragment. The in vitro safety profile is as expected for compounds with similar modes of action. These results suggest that CD123-CODV-TCE may be a promising therapy for patients with relapsed/refractory AML.
Collapse
Affiliation(s)
- Hélène Bonnevaux
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Stephane Guerif
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Jana Albrecht
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Erwan Jouannot
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Thibaud De Gallier
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Christian Beil
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Christian Lange
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Wulf Dirk Leuschner
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Marion Schneider
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Cendrine Lemoine
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Anne Caron
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Céline Amara
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Cédric Barrière
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Justine Siavellis
- Hopitaux Universitaires Paris Ile De France Ouest, Université Versailles Saint Quentin, Paris, France
| | - Valérie Bardet
- Hopitaux Universitaires Paris Ile De France Ouest, Université Versailles Saint Quentin, Paris, France
| | | | | | | | - Ercole Rao
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Peter Wonerow
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Chantal Carrez
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Véronique Blanc
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Karl Hsu
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Dmitri Wiederschain
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Paula G Fraenkel
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| | - Angéla Virone-Oddos
- Sanofi R&D, Vitry-sur-Seine, France; Frankfurt, Germany; and Cambridge, MA, USA
| |
Collapse
|
20
|
Ma XY, Wei L, Lei Z, Chen Y, Ding Z, Chen ZS. Recent progress on targeting leukemia stem cells. Drug Discov Today 2021; 26:1904-1913. [PMID: 34029689 DOI: 10.1016/j.drudis.2021.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/14/2021] [Accepted: 05/17/2021] [Indexed: 10/21/2022]
Abstract
Leukemia is a type of malignant clonal disease of hematopoietic stem cells (HSCs). A small population of leukemic stem cells (LSCs) are responsible for the initiation, drug resistance, and relapse of leukemia. LSCs have the ability to form tumors after xenotransplantation in immunodeficient mice and appear to be common in most human leukemias. Therefore, the eradication of LSCs is an approach with the potential to improve survival or even to cure leukemia. Using recent research in the field of LSCs, we summarize the targeted therapy approaches for the removal of LSCs through surface markers including immune checkpoint molecules, pathways influencing LSC survival, or the survival microenvironment of LSCs. In addition, we introduce the survival microenvironment and survival regulation of LSCs.
Collapse
Affiliation(s)
- Xiang-Yu Ma
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China
| | - Liuya Wei
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| | - Zining Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yanglu Chen
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Gangxing 3rd Rd, High-Tech and Innovation Zone, Jinan, Shandong 250101, PR China
| | - Zhe-Sheng Chen
- School of Pharmacy, Weifang Medical University, Weifang 261053, PR China.
| |
Collapse
|
21
|
Patnaik MM, Mughal TI, Brooks C, Lindsay R, Pemmaraju N. Targeting CD123 in hematologic malignancies: identifying suitable patients for targeted therapy. Leuk Lymphoma 2021; 62:2568-2586. [PMID: 33999767 DOI: 10.1080/10428194.2021.1927021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Following the observation of interleukin 3 receptor α chain (IL-3Rα; CD123) upregulation on leukemia stem cells (LSCs) almost two decades ago, targeted treatment via CD123-diptheria toxin conjugates has now been tested in patients with diverse myeloid malignancies. Targeted eradication of LSCs could result in effective treatments for many challenging diseases initiated by these cells. Consequently, considerable effort has been directed toward targeting CD123 as a potential strategy for treating patients with hematologic malignancies in which CD123 is overexpressed. However, these therapies have had limited success so far, highlighting the need for suitable criteria to identify patients who could benefit from them. Given the diversity in CD123 expression across different hematologic malignancies, understanding CD123 expression patterns and the functional pathogenetic significance is crucial. Here, we review the methodologies available for CD123 assessment and discuss the biological and clinical characteristics of patients for whom CD123-targeting therapies may have a clinical impact.
Collapse
Affiliation(s)
- Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tariq I Mughal
- Division of Hematology-Oncology, Tufts University School of Medicine, Boston, MA, USA.,Research & Clinical Drug Development, Stemline Therapeutics, New York, NY, USA
| | - Christopher Brooks
- Research & Clinical Drug Development, Stemline Therapeutics, New York, NY, USA
| | - Ross Lindsay
- Research & Clinical Drug Development, Stemline Therapeutics, New York, NY, USA
| | - Naveen Pemmaraju
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
22
|
Li Z, Chu X, Gao L, Ling J, Xiao P, Lu J, Wang Y, He H, Li J, Hu Y, Li J, Pan J, Xiao S, Hu S. High Expression of Interleukin-3 Receptor Alpha Chain (CD123) Predicts Favorable Outcome in Pediatric B-Cell Acute Lymphoblastic Leukemia Lacking Prognosis-Defining Genomic Aberrations. Front Oncol 2021; 11:614420. [PMID: 33796456 PMCID: PMC8008053 DOI: 10.3389/fonc.2021.614420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/02/2021] [Indexed: 11/22/2022] Open
Abstract
Background Aberrant expression of CD123 (IL-3Rα) was observed in various hematological malignancies including acute lymphoblastic leukemia (ALL), which is the most common malignancy in childhood. Although widely used for minimal residual disease (MRD) monitoring, the prognostic value of CD123 has not been fully characterized in pediatric B-ALL. This retrospective study aims to evaluate the association between the CD123 expression of leukemic blasts and the outcomes of the pediatric B-ALL patients. Methods A total of 976 pediatric B-ALL, including 328 treated with CCLG-ALL-2008 protocol and 648 treated with CCCG-ALL-2015 protocol, were recruited in this retrospective study. CD123 expression was evaluated by flow cytometry. Patients with >50, 20–50, or <20% of CD123 expressing blasts were grouped into CD123high, CD123low, and CD123neg, respectively. The correlation between CD123 expression and the patients’ clinical characteristics, overall survival (OS), event-free survival (EFS), and relapse-free survival (RFS) were studied statistically. Results Of 976 pediatric B-ALL, 53.4% from the CCLG-ALL-2008 cohort and 49.2% from the CCCG-ALL-2015 cohort were CD123high. In the CCLG-ALL-2008 cohort, CD123high was significantly associated with chromosome hyperdiploidy (p < 0.0001), risk stratification (p = 0.004), and high survival rate (p = 0.005). By comparing clinical outcomes, patients with CD123high displayed favorable prognosis, with a significantly better OS (p = 0.005), EFS (p = 0.017), and RFS (p = 0.045), as compared to patients with CD123low and CD123neg. The prognostic value of CD123 expression was subsequently confirmed in the CCCG-ALL-2015 cohort. Univariate and multivariate cox regression model analysis showed that high CD123 expression was independently associated with favorable EFS (OR: 0.528; 95% CI: 0.327 to 0.853; p = 0.009) in this cohort. In patients without prognosis-defining genomic abnormalities, high CD123 expression strongly indicated superior survival rates and was identified as an independent prognosis factor for EFS and RFS in both cohorts. Conclusions A group of B-ALL lacks prognosis-defining genomic aberrations, which proposes a challenge in risk stratification. Our findings revealed that high CD123 expression of leukemic blasts was associated with favorable clinical outcomes in pediatric B-ALL and CD123 could serve as a promising prognosis predictor, especially in patients without prognosis-defining genetic aberrations.
Collapse
Affiliation(s)
- Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China.,Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Xinran Chu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Li Gao
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jing Ling
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Peifang Xiao
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jun Lu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Yi Wang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Hailong He
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jianqin Li
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Yixin Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jie Li
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, China
| | - Sheng Xiao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Shaoyan Hu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
23
|
Kandeel EZ, Madney Y, Eldin DN, Shafik NF. Overexpression of CD200 and CD123 is a major influential factor in the clinical course of pediatric acute myeloid leukemia. Exp Mol Pathol 2020; 118:104597. [PMID: 33358743 DOI: 10.1016/j.yexmp.2020.104597] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/12/2020] [Accepted: 12/18/2020] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) accounts for approximately 20% of all pediatric acute leukemias. The outcome of AML is still unsatisfactory. CD123 and CD200 were demonstrated to play important roles in hematological malignancies. The aim of this study was to investigate the impact of CD200 and CD123 overexpression and the influence of both proteins on the clinical presentation and disease outcome. Bone marrow (BM) samples from 89 pediatric AML patients were obtained at presentation and after therapy. Cells from the bulk population and from the leukemia stem cell (LSC) compartment were examined by multi parametric flow cytometry. In the bulk population, CD200 was positive in 64/89 (71.9) samples, CD123 was positive in 62/89 (69.7%) samples, and dual CD200 and CD123 positivity was observed in 54/89 (60.7%) samples. CD200/CD123 expressions were observed in LSCs in 64/60 samples respectively (71.9%/67.4%), and co-expressed in 51 samples (57.3%). CD200 was overexpressed in secondary AML (p < 0.05). A multivariate analysis revealed that minimal residual disease (MRD) and lymphadenopathy were associated with CD200 overexpression. Moreover, lymphadenopathy, low platelet count, and MRD were independently associated with CD123 expression. The co-expression of CD200 and CD123 demonstrated a statistically significant relationship with unfavorable cytogenetic karyotypes and high total leucocyte count (TLC). The expression of CD200 and CD123 alone and together had an adverse impact on complete remission (CR), MRD positivity, and overall survival (OS). Cases with MRD on day 28 after induction displayed stable expression patterns of CD200 and CD123. CD200 and CD123 both had a negative influence on clinical presentation and treatment outcome, which remarkably worsened when both were concomitantly overexpressed. CD200 and CD123 can therefore be used as markers of MRD in AML and may also serve as therapeutic targets.
Collapse
Affiliation(s)
- Eman Z Kandeel
- Clinical pathology, National Cancer Institute, Cairo University, Egypt
| | - Youssef Madney
- Pediatric Oncology, National Cancer Institute, Cairo University, Egypt
| | - Dalia Negm Eldin
- Department of Biostatistics and Cancer Epidemiology, National Cancer Institute, Cairo University, Egypt
| | - Nevine F Shafik
- Clinical pathology, National Cancer Institute, Cairo University, Egypt.
| |
Collapse
|
24
|
Chulián S, Martínez-Rubio Á, Pérez-García VM, Rosa M, Blázquez Goñi C, Rodríguez Gutiérrez JF, Hermosín-Ramos L, Molinos Quintana Á, Caballero-Velázquez T, Ramírez-Orellana M, Castillo Robleda A, Fernández-Martínez JL. High-Dimensional Analysis of Single-Cell Flow Cytometry Data Predicts Relapse in Childhood Acute Lymphoblastic Leukaemia. Cancers (Basel) 2020; 13:cancers13010017. [PMID: 33374500 PMCID: PMC7793064 DOI: 10.3390/cancers13010017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/02/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary B-cell Acute Lymphoblastic Leukaemia is one of the most common cancers in childhood, with 20% of patients eventually relapsing. Flow cytometry is routinely used for diagnosis and follow-up, but it currently does not provide prognostic value at diagnosis. The volume and the high-dimensional character of this data makes it ideal for its exploitation by means of Artificial Intelligence methods. We collected flow cytometry data from 56 patients from two hospitals. We analysed differences in intensity of marker expression in order to predict relapse at the moment of diagnosis. We finally correlated this data with biomolecular information, constructing a classifier based on CD38 expression. Abstract Artificial intelligence methods may help in unveiling information that is hidden in high-dimensional oncological data. Flow cytometry studies of haematological malignancies provide quantitative data with the potential to be used for the construction of response biomarkers. Many computational methods from the bioinformatics toolbox can be applied to these data, but they have not been exploited in their full potential in leukaemias, specifically for the case of childhood B-cell Acute Lymphoblastic Leukaemia. In this paper, we analysed flow cytometry data that were obtained at diagnosis from 56 paediatric B-cell Acute Lymphoblastic Leukaemia patients from two local institutions. Our aim was to assess the prognostic potential of immunophenotypical marker expression intensity. We constructed classifiers that are based on the Fisher’s Ratio to quantify differences between patients with relapsing and non-relapsing disease. We also correlated this with genetic information. The main result that arises from the data was the association between subexpression of marker CD38 and the probability of relapse.
Collapse
Affiliation(s)
- Salvador Chulián
- Department of Mathematics, Universidad de Cádiz, Puerto Real, 11510 Cádiz, Spain; (S.C.); (Á.M.-R.); (M.R.)
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain
| | - Álvaro Martínez-Rubio
- Department of Mathematics, Universidad de Cádiz, Puerto Real, 11510 Cádiz, Spain; (S.C.); (Á.M.-R.); (M.R.)
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain
| | - Víctor M. Pérez-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
- Instituto de Matemática Aplicada a la Ciencia y la Ingeniería (IMACI), Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
- ETSI Industriales, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain
- Correspondence:
| | - María Rosa
- Department of Mathematics, Universidad de Cádiz, Puerto Real, 11510 Cádiz, Spain; (S.C.); (Á.M.-R.); (M.R.)
- Biomedical Research and Innovation Institute of Cádiz (INiBICA), Hospital Universitario Puerta del Mar, 11009 Cádiz, Spain
| | - Cristina Blázquez Goñi
- Department of Paediatric Haematology and Oncology, 11407 Hospital de Jerez Cádiz, Spain; (C.B.G.); (J.F.R.G.); (L.H.-R.)
| | | | - Lourdes Hermosín-Ramos
- Department of Paediatric Haematology and Oncology, 11407 Hospital de Jerez Cádiz, Spain; (C.B.G.); (J.F.R.G.); (L.H.-R.)
| | | | | | - Manuel Ramírez-Orellana
- Department of Paediatric Haematology and Oncology, Hospital Infantil Universitario Niño Jesús, Instituto Investigación Sanitaria La Princesa, 28009 Madrid, Spain; (M.R.-O.); (A.C.R.)
| | - Ana Castillo Robleda
- Department of Paediatric Haematology and Oncology, Hospital Infantil Universitario Niño Jesús, Instituto Investigación Sanitaria La Princesa, 28009 Madrid, Spain; (M.R.-O.); (A.C.R.)
| | - Juan Luis Fernández-Martínez
- Department of Mathematics, Group of Inverse Problems, Optimisation and Machine Learning, University of Oviedo, 33005 Oviedo, Spain;
| |
Collapse
|
25
|
El Achi H, Dupont E, Paul S, Khoury JD. CD123 as a Biomarker in Hematolymphoid Malignancies: Principles of Detection and Targeted Therapies. Cancers (Basel) 2020; 12:cancers12113087. [PMID: 33113953 PMCID: PMC7690688 DOI: 10.3390/cancers12113087] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/10/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary CD123 is overexpressed in multiple hematologic malignancies. Advances in CD123-targeted therapies over the past decade have positioned this molecule as an integral biomarker in current practice. This review provides an overview of CD123 biology and in-depth discussion of clinical laboratory techniques used to determine CD123 expression in various hematolymphoid neoplasms. In addition, we describe various pharmacologic strategies and agents that are available or under evaluation for targeting CD123. Abstract CD123, the α chain of the interleukin 3 receptor, is a cytokine receptor that is overexpressed in multiple hematolymphoid neoplasms, including acute myeloid leukemia, blastic plasmacytoid dendritic cell neoplasm, acute lymphoblastic leukemia, hairy cell leukemia, and systemic mastocytosis. Importantly, CD123 expression is upregulated in leukemic stem cells relative to non-neoplastic hematopoietic stem cells, which makes it a useful diagnostic and therapeutic biomarker in hematologic malignancies. Varying levels of evidence have shown that CD123-targeted therapy represents a promising therapeutic approach in several cancers. Tagraxofusp, an anti-CD123 antibody conjugated to a diphtheria toxin, has been approved for use in patients with blastic plasmacytoid dendritic cell neoplasm. Multiple clinical trials are investigating the use of various CD123-targeting agents, including chimeric antigen receptor-modified T cells (expressing CD123, monoclonal antibodies, combined CD3-CD123 dual-affinity retargeting antibody therapy, recombinant fusion proteins, and CD123-engager T cells. In this review, we provide an overview of laboratory techniques used to evaluate and monitor CD123 expression, describe the strengths and limitations of detecting this biomarker in guiding therapy decisions, and provide an overview of the pharmacologic principles and strategies used in CD123-targeted therapies.
Collapse
Affiliation(s)
- Hanadi El Achi
- Department of Pathology and Laboratory Medicine, The University of Texas at Houston, Houston, TX 77030, USA;
| | - Edouard Dupont
- Faculty of Pharmacy of Paris, Paris Descartes University, 75270 Paris, France;
| | - Shilpa Paul
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Joseph D. Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
26
|
Shi M, Su RJ, Parmar KP, Chaudhry R, Sun K, Rao J, Chen M. CD123: A Novel Biomarker for Diagnosis and Treatment of Leukemia. Cardiovasc Hematol Disord Drug Targets 2020; 19:195-204. [PMID: 31244444 DOI: 10.2174/1871529x19666190627100613] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 03/20/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Leukemia is a group of progressive hematologic malignancies derived from stem cells in bone marrow which causes a large number of cancer deaths. Even with treatment such as traditional chemotherapy, targeted therapy, and allogeneic stem cell transplantation (allo-HSCT), many patients suffer from relapse/refractory disease, and the overall survival is dismal. Leukemic stem cells (LSCs) are induced by gene mutations and undergo an aberrant and poorly regulated proliferation process which is involved in the evolution, relapse, and drug-resistance of leukemia. Emerging studies demonstrate that CD123, the interleukin 3 receptor alpha (IL-3Rα), is highly expressed in LSCs, while not normal hematopoietic stem cells (HSCs), and associates with treatment response, minimal residual disease (MRD) detection and prognosis. Furthermore, CD123 is an important marker for the identification and targeting of LSCs for refractory or relapsed leukemia. Anti-CD123 target-therapies in pre-clinical studies and clinical trials confirm the utility of anti-CD123 neutralizing antibody-drugs, CD3×CD123 bispecific antibodies, dual-affinity retargeting (DART), and anti-CD123 chimeric antigen receptor-modified T-cell (CAR-T) therapies in progress. This review summarizes the most recent progress on the study of CD123 biology and the development of novel CD123-targeted therapies.
Collapse
Affiliation(s)
- Mingyue Shi
- Department of Pathology and Laboratory Medicine, UT Southwestern Medical Center, Dallas, TX 75390, United States.,Department of Hematology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Henan, China
| | - Ruijun J Su
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Ronald Reagan UCLA Medical Center, Los Angeles, CA 90095, United States
| | - Kamal-Preet Parmar
- Department of Pathology and Laboratory Medicine, UT Southwestern Medical Center, Dallas, TX 75390, United States
| | - Rahman Chaudhry
- Department of Pathology and Laboratory Medicine, UT Southwestern Medical Center, Dallas, TX 75390, United States
| | - Kai Sun
- Department of Hematology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Henan, China
| | - Jianyu Rao
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Ronald Reagan UCLA Medical Center, Los Angeles, CA 90095, United States
| | - Mingyi Chen
- Department of Pathology and Laboratory Medicine, UT Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
27
|
Sugita M, Guzman ML. CD123 as a Therapeutic Target Against Malignant Stem Cells. Hematol Oncol Clin North Am 2020; 34:553-564. [DOI: 10.1016/j.hoc.2020.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
Clinical Impact of CD25/CD123 Coexpression in Adult B-Cell Acute Lymphoblastic Leukemia Patients. Adv Hematol 2020; 2020:9545717. [PMID: 32518561 PMCID: PMC7256740 DOI: 10.1155/2020/9545717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/19/2020] [Accepted: 04/15/2020] [Indexed: 11/26/2022] Open
Abstract
This study aimed to determine the clinical impact of CD25+/CD123+ coexpression in adult B-cell acute lymphoblastic leukemia (B-ALL) cases. One hundred and twenty newly diagnosed B-ALL patients (≤60 years old) were included in this study. CD123 and CD25 expression on leukemic blast cells were assessed using flow cytometry. CD25+/CD123+ coexpression was detected in 40/120 B-ALL patients (33.3%). All B-ALL patients showed CD25+/CD123+ coexpression had lower induction of remission response and shorter overall survival as compared to B-ALL cases lacking coexpression. In conclusion, CD25+/CD123+ positive coexpression is a reliable flow cytometry marker for prediction of the outcome of adult B-ALL patients and could be used as a novel parameter for risk stratification of adult B-ALL cases.
Collapse
|
29
|
Yang L, Shi P, Zhao G, Xu J, Peng W, Zhang J, Zhang G, Wang X, Dong Z, Chen F, Cui H. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther 2020; 5:8. [PMID: 32296030 PMCID: PMC7005297 DOI: 10.1038/s41392-020-0110-5] [Citation(s) in RCA: 1139] [Impact Index Per Article: 227.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/15/2019] [Accepted: 12/19/2019] [Indexed: 12/18/2022] Open
Abstract
Since cancer stem cells (CSCs) were first identified in leukemia in 1994, they have been considered promising therapeutic targets for cancer therapy. These cells have self-renewal capacity and differentiation potential and contribute to multiple tumor malignancies, such as recurrence, metastasis, heterogeneity, multidrug resistance, and radiation resistance. The biological activities of CSCs are regulated by several pluripotent transcription factors, such as OCT4, Sox2, Nanog, KLF4, and MYC. In addition, many intracellular signaling pathways, such as Wnt, NF-κB (nuclear factor-κB), Notch, Hedgehog, JAK-STAT (Janus kinase/signal transducers and activators of transcription), PI3K/AKT/mTOR (phosphoinositide 3-kinase/AKT/mammalian target of rapamycin), TGF (transforming growth factor)/SMAD, and PPAR (peroxisome proliferator-activated receptor), as well as extracellular factors, such as vascular niches, hypoxia, tumor-associated macrophages, cancer-associated fibroblasts, cancer-associated mesenchymal stem cells, extracellular matrix, and exosomes, have been shown to be very important regulators of CSCs. Molecules, vaccines, antibodies, and CAR-T (chimeric antigen receptor T cell) cells have been developed to specifically target CSCs, and some of these factors are already undergoing clinical trials. This review summarizes the characterization and identification of CSCs, depicts major factors and pathways that regulate CSC development, and discusses potential targeted therapy for CSCs.
Collapse
Affiliation(s)
- Liqun Yang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Pengfei Shi
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Gaichao Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Jie Xu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Wen Peng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Jiayi Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Guanghui Zhang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Xiaowen Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China
| | - Fei Chen
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, 400716, Chongqing, China.
- Cancer Center, Medical Research Institute, Southwest University, 400716, Chongqing, China.
| |
Collapse
|
30
|
Chames P, Wurch T. [Bispecific antibodies, novel therapeutic candidates harnessing the immune system]. Med Sci (Paris) 2020; 35:1072-1082. [PMID: 31903920 DOI: 10.1051/medsci/2019242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Over the past ten years, an increased knowledge of tumor biology and immunology allowed the design and development of novel therapeutic antibody and protein scaffold formats, where bispecific antibodies (Abs) play a major role. The latter molecules can (1) bring novel pharmacological properties through the co-engagement of two targets, (2) increase the safety profile as compared to a combination of two antibodies thanks to a targeted relocation to the tumor and (3) reduce development and manufacturing costs associated with single drug product. This review analyzes the different bispecific antibodies and scaffolds described in the field of immuno-oncology, their structure and major pharmacological and physico-chemical properties.
Collapse
Affiliation(s)
- Patrick Chames
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Thierry Wurch
- Centre d'Innovation Thérapeutique en Oncologie - Servier, F78290 Croissy-sur-Seine, France
| |
Collapse
|
31
|
Mahalleh M, Shabani M, Rayzan E, Rezaei N. Reinforcing the primary immunotherapy modulators against acute leukemia; monoclonal antibodies in AML. Immunotherapy 2019; 11:1583-1600. [PMID: 31841068 DOI: 10.2217/imt-2019-0043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent therapeutic advances in cancer treatment recruit immune system potentiation against malignant cells. Numerous ongoing clinical trials on immunotherapy methods, either monotherapy or combination therapy, are investigating the impeding factors on the way of acute myeloid leukemia (AML) treatment. Due to the genetic diversity in AML progenitors, combining various strategies is more likely to be useful for improving patient outcomes. This review describes the details of applying monoclonal antibodies against AML, focusing on CD33, CD123, FLT3, CD45 and CD66 targeting. Furthermore, it clarifies the importance of immunotoxins, bispecific antibodies, chimeric antigen receptor (CAR)-T cells and T cell receptor-modified cells as reinforcing agents for monoclonal antibodies.
Collapse
Affiliation(s)
- Mehrdad Mahalleh
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsima Shabani
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Elham Rayzan
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Fournier B, Balducci E, Duployez N, Clappier E, Cuccuini W, Arfeuille C, Caye-Eude A, Delabesse E, Bottollier-Lemallaz Colomb E, Nebral K, Chrétien ML, Derrieux C, Cabannes-Hamy A, Dumezy F, Etancelin P, Fenneteau O, Frayfer J, Gourmel A, Loosveld M, Michel G, Nadal N, Penther D, Tigaud I, Fournier E, Reismüller B, Attarbaschi A, Lafage-Pochitaloff M, Baruchel A. B-ALL With t(5;14)(q31;q32); IGH-IL3 Rearrangement and Eosinophilia: A Comprehensive Analysis of a Peculiar IGH-Rearranged B-ALL. Front Oncol 2019; 9:1374. [PMID: 31921638 PMCID: PMC6914849 DOI: 10.3389/fonc.2019.01374] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/21/2019] [Indexed: 02/03/2023] Open
Abstract
Background: B-cell acute lymphoblastic leukemia associated with t(5;14)(q31;q32); IGH-IL3 is an exceptional cause of eosinophilia. The IGH enhancer on 14q32 is juxtaposed to the IL3 gene on 5q31, leading to interleukin-3 overproduction and release of mature eosinophils in the blood. Clinical, biological and outcome data are extremely scarce in the literature. Except for eosinophilia, no relevant common feature has been highlighted in these patients. However, it has been classified as a distinct entity in the World Health Organization classification. Cases Presentation: Eight patients with t(5;14)(q31;q32) treated by French or Austrian protocols were retrospectively enrolled. Array comparative genomic hybridization, multiplex ligation-dependent probe amplification or genomic PCR search for IKZF1 deletion were performed in 7. Sixteen patients found through an exhaustive search in the literature were also analyzed. For those 24 patients, median age at diagnosis is 14.3 years with a male predominance (male to female ratio = 5). Eosinophilia-related symptoms are common (neurologic in 26%, thromboembolic in 26% or pulmonary in 50%). Median white blood cells count is high (72 × 109/L) and linked to eosinophilia (median: 32 × 109/L). Peripheral blasts are present at a low level or absent (median: 0 × 109/L; range: 0–37 × 109/L). Bone marrow morphology is marked by a low blast infiltration (median: 42%). We found an IKZF1 deletion in 5 out of 7 analyzable patients Outcome data are available for 14 patients (median follow-up: 28 months): 8 died and 6 are alive in complete remission. Some of these features are concordant with those seen in patients with other IGH-rearranged B-cell acute lymphoblastic leukemias: young age at onset, male sex, low blast count, high incidence of IKZF1 deletion and intermediate prognosis. Conclusion: Based on shared epidemiological and biological features, B-cell acute lymphoblastic leukemia with t(5;14)(q31;q32) is a peculiar subset of IGH-rearranged B-cell acute lymphoblastic leukemia with an intermediate prognosis and particular clinical features related to eosinophilia.
Collapse
Affiliation(s)
- Benjamin Fournier
- Department of Pediatric Hematology and Immunology, University Hospital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Estelle Balducci
- Hematology Laboratory, University Hospital Paul-Brousse, Assistance Publique des Hôpitaux de Paris (APHP), Villejuif, France
| | | | - Emmanuelle Clappier
- Hematology Laboratory, University Hospital Saint-Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Wendy Cuccuini
- Hematology Laboratory, University Hospital Saint-Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Chloé Arfeuille
- Department of Genetics, University Hospital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Aurélie Caye-Eude
- Department of Genetics, University Hospital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Eric Delabesse
- Department of Haematology, Institut Universitaire de Cancérologie de Toulouse, CHU de Toulouse, Toulouse, France
| | | | - Karin Nebral
- Children's Cancer Research Institute (CCRI), St. Anna Kinderkrebsforschung, Vienna, Austria
| | | | | | - Aurélie Cabannes-Hamy
- Teenagers and Young Adults Hematology Unit, University Hospital Saint-Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Florent Dumezy
- Department of Hematology, University Hospital, Lille, France
| | | | - Odile Fenneteau
- Hematology Laboratory, University Hospital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| | - Jamile Frayfer
- Department of Hematology, Hospital Saint Faron, Meaux, France
| | - Antoine Gourmel
- Department of Pediatric Oncology, Hematology, Immunology, University Hospital of Amiens, Amiens, France
| | - Marie Loosveld
- Hematology Laboratory, Timone Hospital, Assistance Publique-Hôpitaux de Marseille (APHM), CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Aix Marseille University, Marseille, France
| | - Gérard Michel
- Department of Pediatric Hematology, Aix Marseille University, Marseille, France
| | - Nathalie Nadal
- Laboratory of Cytogenetics, University Hospital of Dijon, Dijon, France
| | - Dominique Penther
- Department of Oncology Genetics, Henri Becquerel Center, Rouen, France
| | | | - Elise Fournier
- Department of Hematology, University Hospital, Lille, France
| | - Bettina Reismüller
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Andishe Attarbaschi
- Department of Pediatric Hematology and Oncology, St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Marina Lafage-Pochitaloff
- Hematological Cytogenetics Laboratory, Timone Hospital-Assistance Publique-Hôpitaux de Marseille (AP-HM), Aix-Marseille University, Groupe Francophone de Cytogénétique Hématologique (GFCH), Marseille, France
| | - André Baruchel
- Department of Pediatric Hematology and Immunology, University Hospital Robert Debré, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France.,Institut Universitaire d'Hématologie, EA-3518, University Hospital Saint-Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
| |
Collapse
|
33
|
Dougan M, Dranoff G, Dougan SK. GM-CSF, IL-3, and IL-5 Family of Cytokines: Regulators of Inflammation. Immunity 2019; 50:796-811. [PMID: 30995500 DOI: 10.1016/j.immuni.2019.03.022] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/11/2019] [Accepted: 03/22/2019] [Indexed: 01/27/2023]
Abstract
The β common chain cytokines GM-CSF, IL-3, and IL-5 regulate varied inflammatory responses that promote the rapid clearance of pathogens but also contribute to pathology in chronic inflammation. Therapeutic interventions manipulating these cytokines are approved for use in some cancers as well as allergic and autoimmune disease, and others show promising early clinical activity. These approaches are based on our understanding of the inflammatory roles of these cytokines; however, GM-CSF also participates in the resolution of inflammation, and IL-3 and IL-5 may also have such properties. Here, we review the functions of the β common cytokines in health and disease. We discuss preclinical and clinical data, highlighting the potential inherent in targeting these cytokine pathways, the limitations, and the important gaps in understanding of the basic biology of this cytokine family.
Collapse
Affiliation(s)
- Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Glenn Dranoff
- Novartis Institute for Biomedical Research, Cambridge, MA, USA.
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Sun S, Zou H, Li L, Liu Q, Ding N, Zeng L, Li H, Mao S. CD123/CD33 dual-antibody modified liposomes effectively target acute myeloid leukemia cells and reduce antigen-negative escape. Int J Pharm 2019; 568:118518. [DOI: 10.1016/j.ijpharm.2019.118518] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/30/2019] [Accepted: 07/11/2019] [Indexed: 12/29/2022]
|
35
|
Liu Y, Bewersdorf JP, Stahl M, Zeidan AM. Immunotherapy in acute myeloid leukemia and myelodysplastic syndromes: The dawn of a new era? Blood Rev 2018; 34:67-83. [PMID: 30553527 DOI: 10.1016/j.blre.2018.12.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/02/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Immunotherapy has revolutionized therapy in both solid and liquid malignancies. The ability to cure acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) with an allogeneic hematopoietic stem cell transplant (HSCT) is proof of concept for the application of immunotherapy in AML and MDS. However, outside of HSCT, only the anti-CD33 antibody drug conjugate gemtuzumab ozogamicin is currently approved as an antibody-targeted therapy for AML. Several avenues of immunotherapeutic drugs are currently in different stages of clinical development. Here, we review recent advances in antibody-based therapy, immune checkpoint inhibitors, vaccines and adoptive cell-based therapy for patients with AML and MDS. First, we discuss different antibody constructs. Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein-1 (PD-1) and CD47 as well as peptide, dendritic cell and dendritic/AML cell-based vaccines are reviewed next. Lastly, adoptive cell-based therapy including chimeric antigen receptor (CAR)-T cell and NK cell therapy is discussed.
Collapse
Affiliation(s)
- Yuxin Liu
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Medicine, Section of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
36
|
Emerging functional markers for cancer stem cell-based therapies: Understanding signaling networks for targeting metastasis. Semin Cancer Biol 2018; 53:90-109. [PMID: 29966677 DOI: 10.1016/j.semcancer.2018.06.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 12/18/2022]
Abstract
Metastasis is one of the most challenging issues in cancer patient management, and effective therapies to specifically target disease progression are missing, emphasizing the urgent need for developing novel anti-metastatic therapeutics. Cancer stem cells (CSCs) gained fast attention as a minor population of highly malignant cells within liquid and solid tumors that are responsible for tumor onset, self-renewal, resistance to radio- and chemotherapies, and evasion of immune surveillance accelerating recurrence and metastasis. Recent progress in the identification of their phenotypic and molecular characteristics and interactions with the tumor microenvironment provides great potential for the development of CSC-based targeted therapies and radical improvement in metastasis prevention and cancer patient prognosis. Here, we report on newly uncovered signaling mechanisms controlling CSC's aggressiveness and treatment resistance, and CSC-specific agents and molecular therapeutics, some of which are currently under investigation in clinical trials, gearing towards decisive functional CSC intrinsic or surface markers. One special research focus rests upon subverted regulatory pathways such as insulin-like growth factor 1 receptor signaling and its interactors in metastasis-initiating cell populations directly related to the gain of stem cell- and EMT-associated properties, as well as key components of the E2F transcription factor network regulating metastatic progression, microenvironmental changes, and chemoresistance. In addition, the study provides insight into systems biology tools to establish complex molecular relationships behind the emergence of aggressive phenotypes from high-throughput data that rely on network-based analysis and their use to investigate immune escape mechanisms or predict clinical outcome-relevant CSC receptor signaling signatures. We further propose that customized vector technologies could drastically enhance systemic drug delivery to target sites, and summarize recent progress and remaining challenges. This review integrates available knowledge on CSC biology, computational modeling approaches, molecular targeting strategies, and delivery techniques to envision future clinical therapies designed to conquer metastasis-initiating cells.
Collapse
|
37
|
Schürch CM. Therapeutic Antibodies for Myeloid Neoplasms-Current Developments and Future Directions. Front Oncol 2018; 8:152. [PMID: 29868474 PMCID: PMC5968093 DOI: 10.3389/fonc.2018.00152] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022] Open
Abstract
Therapeutic monoclonal antibodies (mAbs) such as antibody-drug conjugates, ligand-receptor antagonists, immune checkpoint inhibitors and bispecific T cell engagers have shown impressive efficacy in the treatment of multiple human cancers. Numerous therapeutic mAbs that have been developed for myeloid neoplasms, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), are currently investigated in clinical trials. Because AML and MDS originate from malignantly transformed hematopoietic stem/progenitor cells-the so-called leukemic stem cells (LSCs) that are highly resistant to most standard drugs-these malignancies frequently relapse and have a high disease-specific mortality. Therefore, combining standard chemotherapy with antileukemic mAbs that specifically target malignant blasts and particularly LSCs or utilizing mAbs that reinforce antileukemic host immunity holds great promise for improving patient outcomes. This review provides an overview of therapeutic mAbs for AML and MDS. Antibody targets, the molecular mechanisms of action, the efficacy in preclinical leukemia models, and the results of clinical trials are discussed. New developments and future studies of therapeutic mAbs in myeloid neoplasms will advance our understanding of the immunobiology of these diseases and enhance current therapeutic strategies.
Collapse
Affiliation(s)
- Christian M. Schürch
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
38
|
Petrov JC, Wada M, Pinz KG, Yan LE, Chen KH, Shuai X, Liu H, Chen X, Leung LH, Salman H, Hagag N, Liu F, Jiang X, Ma Y. Compound CAR T-cells as a double-pronged approach for treating acute myeloid leukemia. Leukemia 2018. [PMID: 29515236 PMCID: PMC5990523 DOI: 10.1038/s41375-018-0075-3] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Acute myeloid leukemia (AML) bears heterogeneous cells that can consequently offset killing by single-CAR-based therapy, which results in disease relapse. Leukemic stem cells (LSCs) associated with CD123 expression comprise a rare population that also plays an important role in disease progression and relapse. Here, we report on the robust anti-tumor activity of a compound CAR (cCAR) T-cell possessing discrete scFv domains targeting two different AML antigens, CD123, and CD33, simultaneously. We determined that the resulting cCAR T-cells possessed consistent, potent, and directed cytotoxicity against each target antigen population. Using four leukemia mouse models, we found superior in vivo survival after cCAR treatment. We also designed an alemtuzumab safety-switch that allowed for rapid cCAR therapy termination in vivo. These findings indicate that targeting both CD123 and CD33 on AML cells may be an effective strategy for eliminating both AML bulk disease and LSCs, and potentially prevent relapse due to antigen escape or LSC persistence.
Collapse
Affiliation(s)
- Jessica C Petrov
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Masayuki Wada
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA.
| | - Kevin G Pinz
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Lulu E Yan
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Kevin H Chen
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Xiao Shuai
- Department of Hematology West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hua Liu
- Department of Pathology, Stony Brook Medicine, Stony Brook, NY, 11794, USA
| | - Xi Chen
- Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Lai-Han Leung
- Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Huda Salman
- Department of Internal Medicine Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY, 11794, USA
| | - Nabil Hagag
- Department of Internal Medicine Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY, 11794, USA
| | - Fang Liu
- Department of Hematology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Xun Jiang
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA
| | - Yupo Ma
- iCell Gene Therapeutics LLC Research & Development Division Long Island High Technology Incubator, 25 Health Science Drive, Stony Brook, NY, 11790, USA. .,Department of Pathology, Stony Brook Medicine, Stony Brook, NY, 11794, USA. .,Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
39
|
Oliveira E, Bacelar TS, Ciudad J, Ribeiro MCM, Garcia DRN, Sedek L, Maia SF, Aranha DB, Machado IC, Ikeda A, Baglioli BF, Lopez-Duarte N, Teixeira LAC, Szczepanski T, Silva MLM, Land MGP, Orfao A, Costa ES. Altered neutrophil immunophenotypes in childhood B‑cell precursor acute lymphoblastic leukemia. Oncotarget 2017; 7:24664-76. [PMID: 27028865 PMCID: PMC5029732 DOI: 10.18632/oncotarget.8369] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/02/2016] [Indexed: 12/22/2022] Open
Abstract
An increasing number of evidences suggest a genetic predisposition in acute lymphoblastic leukemia (ALL) that might favor the occurrence of the driver genetic alterations. Such genetic background might also translate into phenotypic alterations of residual hematopoietic cells. Whether such phenotypic alterations are present in bone marrow (BM) cells from childhood B-cell precursor (BCP)-ALL remains to be investigated. Here we analyzed the immunophenotypic profile of BM and peripheral blood (PB) maturing/matured neutrophils from 118 children with BCP-ALL and their relationship with the features of the disease. Our results showed altered neutrophil phenotypes in most (77%) BCP-ALL cases. The most frequently altered marker was CD10 (53%), followed by CD33 (34%), CD13 (15%), CD15/CD65 (10%) and CD123 (7%). Of note, patients with altered neutrophil phenotypes had younger age (p = 0.03) and lower percentages of BM maturing neutrophils (p = 0.004) together with greater BM lymphocyte (p = 0.04), and mature B-cell (p = 0.03) counts. No significant association was found between an altered neutrophil phenotype and other disease features. These findings point out the potential existence of an altered residual hematopoiesis in most childhood BCP-ALL cases.
Collapse
Affiliation(s)
- Elen Oliveira
- Clinical Medicine Postgraduate Program, College of Medicine, Rio de Janeiro Federal University (UFRJ), Rio de Janeiro, Brazil.,Cytometry Service, Institute of Pediatrics and Puericulture Martagão Gesteira (IPPMG), UFRJ, Rio de Janeiro, Brazil
| | - Thiago S Bacelar
- Cytometry Service, Institute of Pediatrics and Puericulture Martagão Gesteira (IPPMG), UFRJ, Rio de Janeiro, Brazil
| | - Juana Ciudad
- Departament of Medicine and Cytometry Service, Cancer Research Center (IBMCC, USAL-CSIC), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain
| | | | - Daniela R N Garcia
- Clinical Medicine Postgraduate Program, College of Medicine, Rio de Janeiro Federal University (UFRJ), Rio de Janeiro, Brazil.,Cytogenetics Department, Bone Marrow Transplantation Unit and Oncology Post Graduation Program, National Cancer Institute (INCa), Rio de Janeiro, Brazil
| | - Lukasz Sedek
- Department of Pediatric Hematology/Oncology, Medical University of Silesia, Zabrze, Poland
| | - Simone F Maia
- Service of Pediatric Hematology, Federal Lagoa Hospital (HFL), Rio de Janeiro, Brazil
| | - Daniel B Aranha
- Clinical Medicine Postgraduate Program, College of Medicine, Rio de Janeiro Federal University (UFRJ), Rio de Janeiro, Brazil
| | - Indyara C Machado
- Service of Pediatric Hematology, São José do Avaí Hospital (HSJA), Itaperuna, Rio de Janeiro, Brazil
| | - Arissa Ikeda
- Service of Pediatric Hematology/Oncology, Servidores do Estado Federal Hospital (HSE), Rio de Janeiro, Brazil
| | - Bianca F Baglioli
- Service of Pediatric Hematology, Children's Cancer Hospital of Barretos, Barretos, São Paulo, Brazil
| | - Nathalia Lopez-Duarte
- Cytometry Service, Institute of Pediatrics and Puericulture Martagão Gesteira (IPPMG), UFRJ, Rio de Janeiro, Brazil
| | - Lisandra A C Teixeira
- Clinical Medicine Postgraduate Program, College of Medicine, Rio de Janeiro Federal University (UFRJ), Rio de Janeiro, Brazil.,Cytometry Service, Institute of Pediatrics and Puericulture Martagão Gesteira (IPPMG), UFRJ, Rio de Janeiro, Brazil
| | - Tomasz Szczepanski
- Department of Pediatric Hematology/Oncology, Medical University of Silesia, Zabrze, Poland
| | - Maria Luiza M Silva
- Cytogenetics Department, Bone Marrow Transplantation Unit and Oncology Post Graduation Program, National Cancer Institute (INCa), Rio de Janeiro, Brazil
| | - Marcelo G P Land
- Clinical Medicine Postgraduate Program, College of Medicine, Rio de Janeiro Federal University (UFRJ), Rio de Janeiro, Brazil.,Cytometry Service, Institute of Pediatrics and Puericulture Martagão Gesteira (IPPMG), UFRJ, Rio de Janeiro, Brazil
| | - Alberto Orfao
- Departament of Medicine and Cytometry Service, Cancer Research Center (IBMCC, USAL-CSIC), Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain
| | - Elaine S Costa
- Clinical Medicine Postgraduate Program, College of Medicine, Rio de Janeiro Federal University (UFRJ), Rio de Janeiro, Brazil.,Cytometry Service, Institute of Pediatrics and Puericulture Martagão Gesteira (IPPMG), UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
40
|
Santamaria S, Delgado M, Kremer L, Garcia-Sanz JA. Will a mAb-Based Immunotherapy Directed against Cancer Stem Cells Be Feasible? Front Immunol 2017; 8:1509. [PMID: 29170667 PMCID: PMC5684111 DOI: 10.3389/fimmu.2017.01509] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis suggests that within a tumor, there is a small subpopulation of cells with stem cell properties responsible for tumor maintenance and metastasis generation. This hypothesis also implies that new antitumor drugs, rather than targeting the bulk of the tumor mass, would be more effective if they directly targeted the CSC subpopulation. The CSCs from several types of tumors have been identified with mAbs recognizing surface antigens in these cells; however, antigens specifically or exclusively expressed in the CSC population have not yet been identified. Thus, questioning the possibility of using therapeutic antibodies directed against the CSCs. Here, we review the possibilities of using antibodies directly targeting the CSCs as therapeutic agents in the form of naked antibodies, antibodies conjugated to nanoparticles, or antibody cocktails.
Collapse
Affiliation(s)
- Silvia Santamaria
- Cancer Genetics and Cancer Stem Cell Laboratory, Centro de Investigaciones Biologicas, Department of Cellular and Molecular Medicine, Spanish National Research Council (CSIC), Madrid, Spain
| | - Marisa Delgado
- Cancer Genetics and Cancer Stem Cell Laboratory, Centro de Investigaciones Biologicas, Department of Cellular and Molecular Medicine, Spanish National Research Council (CSIC), Madrid, Spain
| | - Leonor Kremer
- Centro Nacional de Biotecnologia, Department of Immunology and Oncology, Spanish National Research Council (CSIC), Madrid, Spain
| | - Jose A Garcia-Sanz
- Cancer Genetics and Cancer Stem Cell Laboratory, Centro de Investigaciones Biologicas, Department of Cellular and Molecular Medicine, Spanish National Research Council (CSIC), Madrid, Spain
| |
Collapse
|
41
|
Wu H, Wang M, Dai B, Zhang Y, Yang Y, Li Q, Duan M, Zhang X, Wang X, Li A, Zhang L. Novel CD123-aptamer-originated targeted drug trains for selectively delivering cytotoxic agent to tumor cells in acute myeloid leukemia theranostics. Drug Deliv 2017; 24:1216-1229. [PMID: 28845698 PMCID: PMC8241133 DOI: 10.1080/10717544.2017.1367976] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022] Open
Abstract
Since conventional chemotherapy for acute myeloid leukemia (AML) has its limitations, a theranostic platform with targeted and efficient drug transport is in demand. In this study, we developed the first CD123 (AML tumor marker) aptamers and designed a novel CD123-aptamer-mediated targeted drug train (TDT) with effective, economical, biocompatible and high drug-loading capacity. These two CD123 aptamers (termed as ZW25 and CY30, respectively) can bind to a CD123 peptide epitope and CD123 + AML cells with high specificities and KD of 29.41 nM and 15.38 nM, respectively, while has minimal cross reactivities to albumin, IgG and trypsin. Further, TDT is self-assembled from two short primers by ligand-modified ZW25 that acted as initiation position for elongation, while intercalated by doxorubicin (Dox). TDT is capable of transporting high capacity of Dox to CD123 + cells and retains the efficacy of Dox, while significantly reducing drug uptake and eased toxicity to CD123- cells in vitro (p < .01). Moreover, TDT can ease Dox cytoxicity to normal tissues, prolong survivals and inhibit tumor growth of mouse xenograft tumor model in vivo. These suggest that CD123 aptamer and CD123 aptamer-mediated targeted drug delivery system may have potential applications for selective delivery cytotoxic agents to CD123-expressing tumors in AML theranostics.
Collapse
Affiliation(s)
- Haibin Wu
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
- Key laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, People’s Republic of China
| | - Meng Wang
- Department of Orthopedics, The No.11 Hospital of PLA, YiNing, XinJiang, People’s Republic of China
| | - Bo Dai
- Shaanxi Center for Stem Cell Application Engineering Research, Xi’an, Shaanxi, People’s Republic of China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, Shaanxi, People’s Republic of China
| | - Yanmin Zhang
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Ying Yang
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Qiao Li
- Clinical Laboratory, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Mingyue Duan
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Xi Zhang
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Xiaomei Wang
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Anmao Li
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
| | - Liyu Zhang
- Shaanxi Institute of Pediatric Diseases, Xi’an Children’s Hospital, Xi’an, Shaanxi, People’s Republic of China
- Key laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
42
|
Audehm S, Krackhardt AM. Specific Adoptive Cellular Immunotherapy in Allogeneic Stem Cell Transplantation. Oncol Res Treat 2017; 40:691-696. [PMID: 29069663 DOI: 10.1159/000484051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents a treatment option for a diversity of advanced hematopoietic malignancies providing hope for long-term responses especially due to immunogenic effects associated with the treatment modality. Despite respectable progress in the field, relapses and/or opportunistic infections are major reasons for the high treatment-related mortality. However, a number of novel immunotherapeutic approaches using defined cell populations have been developed to directly target residual malignant cells as well as defined infectious diseases. We here provide an overview of current adoptive cellular immunotherapies in the context of allo-HSCT and close with an outlook on new directions within the field.
Collapse
Affiliation(s)
- Stefan Audehm
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | |
Collapse
|
43
|
Ruella M, Klichinsky M, Kenderian SS, Shestova O, Ziober A, Kraft DO, Feldman M, Wasik MA, June CH, Gill S. Overcoming the Immunosuppressive Tumor Microenvironment of Hodgkin Lymphoma Using Chimeric Antigen Receptor T Cells. Cancer Discov 2017; 7:1154-1167. [PMID: 28576927 PMCID: PMC5628114 DOI: 10.1158/2159-8290.cd-16-0850] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 04/19/2017] [Accepted: 05/31/2017] [Indexed: 01/03/2023]
Abstract
Patients with otherwise treatment-resistant Hodgkin lymphoma could benefit from chimeric antigen receptor T-cell (CART) therapy. However, Hodgkin lymphoma lacks CD19 and contains a highly immunosuppressive tumor microenvironment (TME). We hypothesized that in Hodgkin lymphoma, CART should target both malignant cells and the TME. We demonstrated CD123 on both Hodgkin lymphoma cells and TME, including tumor-associated macrophages (TAM). In vitro, Hodgkin lymphoma cells convert macrophages toward immunosuppressive TAMs that inhibit T-cell proliferation. In contrast, anti-CD123 CART recognized and killed TAMs, thus overcoming immunosuppression. Finally, we showed in immunodeficient mouse models that CART123 eradicated Hodgkin lymphoma and established long-term immune memory. A novel platform that targets malignant cells and the microenvironment may be needed to successfully treat malignancies with an immunosuppressive milieu.Significance: Anti-CD123 chimeric antigen receptor T cells target both the malignant cells and TAMs in Hodgkin lymphoma, thereby eliminating an important immunosuppressive component of the tumor microenvironment. Cancer Discov; 7(10); 1154-67. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1047.
Collapse
Affiliation(s)
- Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael Klichinsky
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saad S Kenderian
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Olga Shestova
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy Ziober
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel O Kraft
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mariusz A Wasik
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Saar Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
44
|
Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: How to strike the evil at its root. Adv Drug Deliv Rev 2017; 120:89-107. [PMID: 28736304 DOI: 10.1016/j.addr.2017.07.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/10/2017] [Accepted: 07/16/2017] [Indexed: 12/18/2022]
Abstract
Cancer progression to metastatic stages is still unmanageable and the promise of effective anti-metastatic therapy remains largely unmet, emphasizing the need to develop novel therapeutics. The special focus here is on cancer stem cells (CSC) as the seed of tumor initiation, epithelial-mesenchymal transition, chemoresistance and, as a consequence, drivers of metastatic dissemination. We report on targeted therapies gearing towards the CSC's internal and membrane-anchored markers using agents such as antibody derivatives, nucleic therapeutics, small molecules and genetic payloads. Another emphasis lies on novel proceedings envisaged to deliver current and prospective therapies to the target sites using newest viral and non-viral vector technologies. In this review, we summarize recent progress and remaining challenges in therapeutic strategies to combat CSC.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany.
| | - Manish Solanki
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| | - Ottmar Herchenröder
- Institute of Experimental Gene Therapy and Cancer Research, Biomedical Research Center (BMFZ), Rostock University Medical School, Germany
| |
Collapse
|
45
|
A novel immunoliposome mediated by CD123 antibody targeting to acute myeloid leukemia cells. Int J Pharm 2017; 529:531-542. [DOI: 10.1016/j.ijpharm.2017.06.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/16/2017] [Accepted: 06/01/2017] [Indexed: 01/11/2023]
|
46
|
Chen Y, Anastassiadis K, Kranz A, Stewart AF, Arndt K, Waskow C, Yokoyama A, Jones K, Neff T, Lee Y, Ernst P. MLL2, Not MLL1, Plays a Major Role in Sustaining MLL-Rearranged Acute Myeloid Leukemia. Cancer Cell 2017; 31:755-770.e6. [PMID: 28609655 PMCID: PMC5598468 DOI: 10.1016/j.ccell.2017.05.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 03/09/2017] [Accepted: 05/05/2017] [Indexed: 01/11/2023]
Abstract
The MLL1 histone methyltransferase gene undergoes many distinct chromosomal rearrangements to yield poor-prognosis leukemia. The remaining wild-type allele is most commonly, but not always, retained. To what extent the wild-type allele contributes to leukemogenesis is unclear. Here we show, using rigorous, independent animal models, that endogenous MLL1 is dispensable for MLL-rearranged leukemia. Potential redundancy was addressed by co-deleting the closest paralog, Mll2. Surprisingly, Mll2 deletion alone had a significant impact on survival of MLL-AF9-transformed cells, and additional Mll1 loss further reduced viability and proliferation. We show that MLL1/MLL2 collaboration is not through redundancy, but regulation of distinct pathways. These findings highlight the relevance of MLL2 as a drug target in MLL-rearranged leukemia and suggest its broader significance in AML.
Collapse
Affiliation(s)
- Yufei Chen
- Department of Pediatrics, Section of Hematology/Oncology/BMT, University of Colorado, Denver/Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Konstantinos Anastassiadis
- Genomics and Stem Cell Engineering, Biotechnology Center, Technische Universität Dresden, BioInnovations Zentrum, Tatzberg 47, Dresden 01307, Germany
| | - Andrea Kranz
- Genomics and Stem Cell Engineering, Biotechnology Center, Technische Universität Dresden, BioInnovations Zentrum, Tatzberg 47, Dresden 01307, Germany
| | - A Francis Stewart
- Genomics and Stem Cell Engineering, Biotechnology Center, Technische Universität Dresden, BioInnovations Zentrum, Tatzberg 47, Dresden 01307, Germany
| | - Kathrin Arndt
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Claudia Waskow
- Regeneration in Hematopoiesis and Animal Models in Hematopoiesis, Institute for Immunology, Medical Faculty, Technische Universität Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| | - Akihiko Yokoyama
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Mizukami 246-2, Kakuganji, Tsuruoka, Yamagata 997-0052, Japan
| | - Kenneth Jones
- Department of Pediatrics, Section of Hematology/Oncology/BMT, University of Colorado, Denver/Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Tobias Neff
- Department of Pediatrics, Section of Hematology/Oncology/BMT, University of Colorado, Denver/Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Yoo Lee
- Department of Pediatrics, Section of Hematology/Oncology/BMT, University of Colorado, Denver/Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Patricia Ernst
- Department of Pediatrics, Section of Hematology/Oncology/BMT, University of Colorado, Denver/Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado, Denver/Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
47
|
Hoseini SS, Cheung NK. Acute myeloid leukemia targets for bispecific antibodies. Blood Cancer J 2017; 7:e522. [PMID: 28157217 PMCID: PMC5386336 DOI: 10.1038/bcj.2017.2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022] Open
Abstract
Despite substantial gains in our understanding of the genomics of acute myelogenous leukemia (AML), patient survival remains unsatisfactory especially among the older age group. T cell-based therapy of lymphoblastic leukemia is rapidly advancing; however, its application in AML is still lagging behind. Bispecific antibodies can redirect polyclonal effector cells to engage chosen targets on leukemia blasts. When the effector cells are natural-killer cells, both antibody-dependent and antibody-independent mechanisms could be exploited. When the effectors are T cells, direct tumor cytotoxicity can be engaged followed by a potential vaccination effect. In this review, we summarize the AML-associated tumor targets and the bispecific antibodies that have been studied. The potentials and limitations of each of these systems will be discussed.
Collapse
Affiliation(s)
- S S Hoseini
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - N K Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
48
|
Castelli G, Pelosi E, Testa U. Targeted therapies in the treatment of adult acute myeloid leukemias: current status and future perspectives. Int J Hematol Oncol 2016; 5:143-164. [PMID: 30302215 PMCID: PMC6172000 DOI: 10.2217/ijh-2016-0011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 11/29/2016] [Indexed: 12/19/2022] Open
Abstract
The rapid advancement of next-generation sequencing techniques and the identification of molecular driver events responsible for leukemia development are opening the door to new pharmacologic-targeted agents to tailor treatment of acute myeloid leukemia (AML) in individual patients. However, the use of targeted therapies in AML has met with only modest success. Molecular studies have identified AML subsets characterized by driver mutational events, such as NPM1, FLT3-ITD and IDH1-2 mutations, and have provided preclinical evidence that the targeting of these mutant molecules could represent a valuable therapeutic strategy. Recent studies have provided the first pieces of evidence that FLT3 targeting in FLT3-mutant AMLs, IDH1/2 inhibition in IDH-mutant AMLs and targeting membrane molecules preferentially expressed on leukemic progenitor/stem cells, such as CD33 and CD123, represent a clinically valuable strategy.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Hematology, Oncology & Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Elvira Pelosi
- Department of Hematology, Oncology & Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| | - Ugo Testa
- Department of Hematology, Oncology & Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome 00161, Italy
| |
Collapse
|
49
|
Moradi-Kalbolandi S, Habibi-Anbouhi M, Golkar M, Behdani M, Rezaei G, Ghazizadeh L, Abolhassani M, Shokrgozar MA. Development of a novel engineered antibody targeting human CD123. Anal Biochem 2016; 511:27-30. [DOI: 10.1016/j.ab.2016.04.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/20/2016] [Accepted: 04/21/2016] [Indexed: 12/20/2022]
|
50
|
Ruella M, Barrett DM, Kenderian SS, Shestova O, Hofmann TJ, Perazzelli J, Klichinsky M, Aikawa V, Nazimuddin F, Kozlowski M, Scholler J, Lacey SF, Melenhorst JJ, Morrissette JJD, Christian DA, Hunter CA, Kalos M, Porter DL, June CH, Grupp SA, Gill S. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J Clin Invest 2016; 126:3814-3826. [PMID: 27571406 DOI: 10.1172/jci87366] [Citation(s) in RCA: 454] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/14/2016] [Indexed: 12/21/2022] Open
Abstract
Potent CD19-directed immunotherapies, such as chimeric antigen receptor T cells (CART) and blinatumomab, have drastically changed the outcome of patients with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL). However, CD19-negative relapses have emerged as a major problem that is observed in approximately 30% of treated patients. Developing approaches to preventing and treating antigen-loss escapes would therefore represent a vertical advance in the field. Here, we found that in primary patient samples, the IL-3 receptor α chain CD123 was highly expressed on leukemia-initiating cells and CD19-negative blasts in bulk B-ALL at baseline and at relapse after CART19 administration. Using intravital imaging in an antigen-loss CD19-negative relapse xenograft model, we determined that CART123, but not CART19, recognized leukemic blasts, established protracted synapses, and eradicated CD19-negative leukemia, leading to prolonged survival. Furthermore, combining CART19 and CART123 prevented antigen-loss relapses in xenograft models. Finally, we devised a dual CAR-expressing construct that combined CD19- and CD123-mediated T cell activation and demonstrated that it provides superior in vivo activity against B-ALL compared with single-expressing CART or pooled combination CART. In conclusion, these findings indicate that targeting CD19 and CD123 on leukemic blasts represents an effective strategy for treating and preventing antigen-loss relapses occurring after CD19-directed therapies.
Collapse
|