1
|
Soleimani Samarkhazan H, Zehtabcheh S, Seraji HR, Beqaj SH, Tayefeh S, Mohammadi MH, Aghaei M. Unveiling the potential of CLL-1: a promising target for AML therapy. Biomark Res 2025; 13:28. [PMID: 39940055 PMCID: PMC11823018 DOI: 10.1186/s40364-025-00738-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Acute myeloid leukemia (AML) remains a formidable blood cancer, despite recent advances in treatment. A significant challenge persists in improving patient outcomes, particularly in addressing relapse and treatment resistance. Identifying new therapeutic targets is critical for advancing AML therapy. C-type lectin-like molecule-1 (CLL-1) has emerged as a promising therapeutic target in AML. This cell surface receptor is highly expressed on AML blasts and demonstrates stable expression throughout disease progression. CLL-1's consistent presence makes it an ideal candidate for monitoring minimal residual disease (MRD), which is a critical indicator for predicting relapse. Beyond its utility as a diagnostic marker, CLL-1 offers exciting potential in the development of immunotherapies. Emerging strategies, such as CAR-T-cell therapy and antibody-drug conjugates (ADCs), are being investigated to leverage the immune system against CLL-1-expressing AML cells. This review examines the structure, function, and expression patterns of CLL-1 in AML and other hematologic malignancies, providing insights into its role in disease pathogenesis and treatment potential. Exploring CLL-1 as a target for diagnosis, MRD monitoring, and immunotherapy opens new avenues for AML treatment. A deeper understanding of its relationship with AML pathogenesis will aid in the development of targeted therapies, offering hope for improved patient outcomes in the future.
Collapse
Affiliation(s)
- Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Zehtabcheh
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamideh Rahmani Seraji
- Department of Hematology and Oncology, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shamim Tayefeh
- UCLA Immunogenetics Center, Immunogenetics (UIC), 1000 Veteran Ave, Los Angeles, CA, 90024, USA
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
Cai Q, Lan H, Yi D, Xian B, Zidan L, Li J, Liao Z. Flow cytometry in acute myeloid leukemia and detection of minimal residual disease. Clin Chim Acta 2025; 564:119945. [PMID: 39209245 DOI: 10.1016/j.cca.2024.119945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Acute myeloid leukemia (AML) is a common type of acute leukemia (AL), belonging to malignant tumors of the hematopoietic system with the characteristics of rapid disease development, control with extreme difficulties, easy recurrence, poor prognosis, and incidence rate increasing with age. The traditionally diagnostic standard of French American British (FAB), being based on the morphological examination with high human subjectivity, can no longer meet the demand of clinical diagnosis and treatment of AML. Requirements of objective accuracy and low-dose sample, have become the indispensable method for AML diagnosis and monitoring prognosis. Flow cytometry is a modern technology that can quickly and accurately detect the series, antigen distribution, differentiation stage of AML cells, minimal residual lesions after AML therapy, so as to provide the great significance in guiding clinical diagnosis, hierarchical treatment, and prognosis judgement. This article will systematically elaborate on the application of flow cytometry in the diagnosis and classification of AML, and the detection of minimal residual lesions, thereby providing reference significance for dynamic monitoring and prognostic observation of AML with different immune subtypes of FAB.
Collapse
Affiliation(s)
- Qihui Cai
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Haiqiang Lan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Deng Yi
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Bojun Xian
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Luo Zidan
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Jianqiao Li
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Zhaohong Liao
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan, Guangdong 528000, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
3
|
de Groot AP, de Haan G. How CBX proteins regulate normal and leukemic blood cells. FEBS Lett 2024; 598:2788-2806. [PMID: 38426219 PMCID: PMC11586599 DOI: 10.1002/1873-3468.14839] [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: 10/19/2023] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Hematopoietic stem cell (HSC) fate decisions are dictated by epigenetic landscapes. The Polycomb Repressive Complex 1 (PRC1) represses genes that induce differentiation, thereby maintaining HSC self-renewal. Depending on which chromobox (CBX) protein (CBX2, CBX4, CBX6, CBX7, or CBX8) is part of the PRC1 complex, HSC fate decisions differ. Here, we review how this occurs. We describe how CBX proteins dictate age-related changes in HSCs and stimulate oncogenic HSC fate decisions, either as canonical PRC1 members or by alternative interactions, including non-epigenetic regulation. CBX2, CBX7, and CBX8 enhance leukemia progression. To target, reprogram, and kill leukemic cells, we suggest and describe multiple therapeutic strategies to interfere with the epigenetic functions of oncogenic CBX proteins. Future studies should clarify to what extent the non-epigenetic function of cytoplasmic CBX proteins is important for normal, aged, and leukemic blood cells.
Collapse
Affiliation(s)
- Anne P. de Groot
- European Research Institute for Biology of Ageing (ERIBA)University Medical Center Groningen (UMCG)The Netherlands
- Sanquin Research, Landsteiner LaboratorySanquin Blood SupplyAmsterdamThe Netherlands
| | - Gerald de Haan
- European Research Institute for Biology of Ageing (ERIBA)University Medical Center Groningen (UMCG)The Netherlands
- Sanquin Research, Landsteiner LaboratorySanquin Blood SupplyAmsterdamThe Netherlands
- Department of Hematology, Amsterdam UMCUniversity of AmsterdamThe Netherlands
| |
Collapse
|
4
|
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
|
5
|
Nair R, Salinas-Illarena A, Sponheimer M, Wullkopf I, Schreiber Y, Côrte-Real JV, Del Pozo Ben A, Marterer H, Thomas D, Geisslinger G, Cinatl J, Subklewe M, Baldauf HM. Novel Vpx virus-like particles to improve cytarabine treatment response against acute myeloid leukemia. Clin Exp Med 2024; 24:155. [PMID: 39003408 PMCID: PMC11246277 DOI: 10.1007/s10238-024-01425-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024]
Abstract
Knowledge of the molecular pathogenesis of acute myeloid leukemia has advanced in recent years. Despite novel treatment options, acute myeloid leukemia remains a survival challenge for elderly patients. We have recently shown that the triphosphohydrolase SAMHD1 is one of the factors determining resistance to Ara-C treatment. Here, we designed and tested novel and simpler virus-like particles incorporating the lentiviral protein Vpx to efficiently and transiently degrade SAMHD1 and increase the efficacy of Ara-C treatment. The addition of minute amounts of lentiviral Rev protein during production enhanced the generation of virus-like particles. In addition, we found that our 2nd generation of virus-like particles efficiently targeted and degraded SAMHD1 in AML cell lines with high levels of SAMHD1, thereby increasing Ara-CTP levels and response to Ara-C treatment. Primary AML blasts were generally less responsive to VLP treatment. In summary, we have been able to generate novel and simpler virus-like particles that can efficiently deliver Vpx to target cells.
Collapse
Affiliation(s)
- Ramya Nair
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany
| | - Alejandro Salinas-Illarena
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany
| | - Monika Sponheimer
- Department of Medicine III, University Hospital, LMU, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Inès Wullkopf
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany
| | - Yannick Schreiber
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596, Frankfurt Am Main, Germany
| | - João Vasco Côrte-Real
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany
- CIBIO-InBIO, Research Center in Biodiversity and Genetic Resources, University of Porto, 4485-661, Vairão, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, 4169-007, Porto, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, 4485-661, Vairão, Portugal
| | - Augusto Del Pozo Ben
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany
| | - Helena Marterer
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany
| | - Dominique Thomas
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596, Frankfurt Am Main, Germany
- Institute for Clinical Pharmacology, Goethe University Frankfurt, 60590, Frankfurt Am Main, Germany
| | - Gerd Geisslinger
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596, Frankfurt Am Main, Germany
- Institute for Clinical Pharmacology, Goethe University Frankfurt, 60590, Frankfurt Am Main, Germany
| | - Jindrich Cinatl
- Institute for Medical Virology, University Hospital, Goethe University, Frankfurt Am Main, Germany
- Dr. Petra Joh-Forschungshaus, Frankfurt Am Main, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, LMU, Munich, Germany
- Laboratory for Translational Cancer Immunology, LMU Gene Center, Munich, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hanna-Mari Baldauf
- Max Von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Faculty of Medicine, LMU München, Feodor-Lynen-Str. 23, 81377, Munich, Germany.
| |
Collapse
|
6
|
Wu S, Tan Y, Li F, Han Y, Zhang S, Lin X. CD44: a cancer stem cell marker and therapeutic target in leukemia treatment. Front Immunol 2024; 15:1354992. [PMID: 38736891 PMCID: PMC11082360 DOI: 10.3389/fimmu.2024.1354992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
CD44 is a ubiquitous leukocyte adhesion molecule involved in cell-cell interaction, cell adhesion, migration, homing and differentiation. CD44 can mediate the interaction between leukemic stem cells and the surrounding extracellular matrix, thereby inducing a cascade of signaling pathways to regulate their various behaviors. In this review, we focus on the impact of CD44s/CD44v as biomarkers in leukemia development and discuss the current research and prospects for CD44-related interventions in clinical application.
Collapse
Affiliation(s)
- Shuang Wu
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yicheng Tan
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
| | - Fanfan Li
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yixiang Han
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Central Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shenghui Zhang
- Laboratory Animal Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Hematology, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Wenzhou Key laboratory of Hematology, Wenzhou, Zhejiang, China
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaofei Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
7
|
Guo Y, Li X, Xie Y, Wang Y. What influences the activity of Degrader-Antibody conjugates (DACs). Eur J Med Chem 2024; 268:116216. [PMID: 38387330 DOI: 10.1016/j.ejmech.2024.116216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
The targeted protein degradation (TPD) technology employing proteolysis-targeting chimeras (PROTACs) has been widely applied in drug chemistry and chemical biology for the treatment of cancer and other diseases. PROTACs have demonstrated significant advantages in targeting undruggable targets and overcoming drug resistance. However, despite the efficient degradation of targeted proteins achieved by PROTACs, they still face challenges related to selectivity between normal and cancer cells, as well as issues with poor membrane permeability due to their substantial molecular weight. Additionally, the noteworthy toxicity resulting from off-target effects also needs to be addressed. To solve these issues, Degrader-Antibody Conjugates (DACs) have been developed, leveraging the targeting and internalization capabilities of antibodies. In this review, we elucidates the characteristics and distinctions between DACs, and traditional Antibody-drug conjugates (ADCs). Meanwhile, we emphasizes the significance of DACs in facilitating the delivery of PROTACs and delves into the impact of various components on DAC activity. These components include antibody targets, drug-antibody ratio (DAR), linker types, PROTACs targets, PROTACs connections, and E3 ligase ligands. The review also explores the suitability of different targets (antibody targets or PROTACs targets) for DACs, providing insights to guide the design of PROTACs better suited for antibody conjugation.
Collapse
Affiliation(s)
- Yaolin Guo
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yang Xie
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, Sichuan, China.
| |
Collapse
|
8
|
Guarnera L, Bravo-Perez C, Visconte V. Immunotherapy in Acute Myeloid Leukemia: A Literature Review of Emerging Strategies. Bioengineering (Basel) 2023; 10:1228. [PMID: 37892958 PMCID: PMC10604866 DOI: 10.3390/bioengineering10101228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
In the last twenty years, we have witnessed a paradigm shift in the treatment and prognosis of acute myeloid leukemia (AML), thanks to the introduction of new efficient drugs or approaches to refine old therapies, such as Gemtuzumab Ozogamicin, CPX 3-5-1, hypomethylating agents, and Venetoclax, the optimization of conditioning regimens in allogeneic hematopoietic stem cell transplantation and the improvement of supportive care. However, the long-term survival of non-M3 and non-core binding factor-AML is still dismal. For this reason, the expectations for the recently developed immunotherapies, such as antibody-based therapy, checkpoint inhibitors, and chimeric antigen receptor strategies, successfully tested in other hematologic malignancies, were very high. The inherent characteristics of AML blasts hampered the development of these treatments, and the path of immunotherapy in AML has been bumpy. Herein, we provide a detailed review of potential antigenic targets, available data from pre-clinical and clinical trials, and future directions of immunotherapies in AML.
Collapse
Affiliation(s)
- Luca Guarnera
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlos Bravo-Perez
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER—Instituto de Salud Carlos III, 30005 Murcia, Spain
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
| |
Collapse
|
9
|
Molica M, Perrone S, Andriola C, Rossi M. Immunotherapy with Monoclonal Antibodies for Acute Myeloid Leukemia: A Work in Progress. Cancers (Basel) 2023; 15:5060. [PMID: 37894427 PMCID: PMC10605302 DOI: 10.3390/cancers15205060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
In the last few years, molecularly targeted agents and immune-based treatments (ITs) have significantly changed the landscape of anti-cancer therapy. Indeed, ITs have been proven to be very effective when used against metastatic solid tumors, for which outcomes are extremely poor when using standard approaches. Such a scenario has only been partially reproduced in hematologic malignancies. In the context of acute myeloid leukemia (AML), as innovative drugs are eagerly awaited in the relapsed/refractory setting, different ITs have been explored, but the results are still unsatisfactory. In this work, we will discuss the most important clinical studies to date that adopt ITs in AML, providing the basis to understand how this approach, although still in its infancy, may represent a promising therapeutic tool for the future treatment of AML patients.
Collapse
Affiliation(s)
- Matteo Molica
- Department of Hematology-Oncology, Azienda Universitaria Ospedaliera Renato Dulbecco, 88100 Catanzaro, Italy;
| | - Salvatore Perrone
- Department of Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, 04100 Latina, Italy;
| | - Costanza Andriola
- Hematology, Department of Translational and Precision Medicine, Sapienza University, 00100 Rome, Italy;
| | - Marco Rossi
- Department of Hematology-Oncology, Azienda Universitaria Ospedaliera Renato Dulbecco, 88100 Catanzaro, Italy;
| |
Collapse
|
10
|
Bakhtiyari M, Liaghat M, Aziziyan F, Shapourian H, Yahyazadeh S, Alipour M, Shahveh S, Maleki-Sheikhabadi F, Halimi H, Forghaniesfidvajani R, Zalpoor H, Nabi-Afjadi M, Pornour M. The role of bone marrow microenvironment (BMM) cells in acute myeloid leukemia (AML) progression: immune checkpoints, metabolic checkpoints, and signaling pathways. Cell Commun Signal 2023; 21:252. [PMID: 37735675 PMCID: PMC10512514 DOI: 10.1186/s12964-023-01282-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/17/2023] [Indexed: 09/23/2023] Open
Abstract
Acute myeloid leukemia (AML) comprises a multifarious and heterogeneous array of illnesses characterized by the anomalous proliferation of myeloid cells in the bone marrow microenvironment (BMM). The BMM plays a pivotal role in promoting AML progression, angiogenesis, and metastasis. The immune checkpoints (ICs) and metabolic processes are the key players in this process. In this review, we delineate the metabolic and immune checkpoint characteristics of the AML BMM, with a focus on the roles of BMM cells e.g. tumor-associated macrophages, natural killer cells, dendritic cells, metabolic profiles and related signaling pathways. We also discuss the signaling pathways stimulated in AML cells by BMM factors that lead to AML progression. We then delve into the roles of immune checkpoints in AML angiogenesis, metastasis, and cell proliferation, including co-stimulatory and inhibitory ICs. Lastly, we discuss the potential therapeutic approaches and future directions for AML treatment, emphasizing the potential of targeting metabolic and immune checkpoints in AML BMM as prognostic and therapeutic targets. In conclusion, the modulation of these processes through the use of directed drugs opens up new promising avenues in combating AML. Thereby, a comprehensive elucidation of the significance of these AML BMM cells' metabolic and immune checkpoints and signaling pathways on leukemic cells can be undertaken in the future investigations. Additionally, these checkpoints and cells should be considered plausible multi-targeted therapies for AML in combination with other conventional treatments in AML. Video Abstract.
Collapse
Affiliation(s)
- Maryam Bakhtiyari
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Mahsa Liaghat
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hooriyeh Shapourian
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sheida Yahyazadeh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Alipour
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Shaghayegh Shahveh
- American Association of Naturopath Physician (AANP), Washington, DC, USA
| | - Fahimeh Maleki-Sheikhabadi
- Department of Hematology and Blood Banking, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Halimi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Forghaniesfidvajani
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Majid Pornour
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, USA.
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland, USA.
| |
Collapse
|
11
|
Damiani D, Tiribelli M. Checkpoint Inhibitors in Acute Myeloid Leukemia. Biomedicines 2023; 11:1724. [PMID: 37371818 PMCID: PMC10295997 DOI: 10.3390/biomedicines11061724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The prognosis of acute myeloid leukemia (AML) remains unsatisfactory. Among the reasons for the poor response to therapy and high incidence of relapse, there is tumor cell immune escape, as AML blasts can negatively influence various components of the immune system, mostly weakening T-cells. Since leukemic cells can dysregulate immune checkpoints (ICs), receptor-based signal transductors that lead to the negative regulation of T-cells and, eventually, to immune surveillance escape, the inhibition of ICs is a promising therapeutic strategy and has led to the development of so-called immune checkpoint inhibitors (ICIs). ICIs, in combination with conventional chemotherapy, hypomethylating agents or targeted therapies, are being increasingly tested in cases of AML, but the results reported are often conflicting. Here, we review the main issues concerning the immune system in AML, the main pathways leading to immune escape and the results obtained from clinical trials of ICIs, alone or in combination, in newly diagnosed or relapsed/refractory AML.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, Udine Hospital, 33100 Udine, Italy;
- Department of Medicine, Udine University, 33100 Udine, Italy
| |
Collapse
|
12
|
Morris VS, Ghazi H, Fletcher DM, Guinn BA. A Direct Comparison, and Prioritisation, of the Immunotherapeutic Targets Expressed by Adult and Paediatric Acute Myeloid Leukaemia Cells: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:9667. [PMID: 37298623 PMCID: PMC10253696 DOI: 10.3390/ijms24119667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Acute myeloid leukaemia (AML) is characterized by impaired myeloid differentiation resulting in an accumulation of immature blasts in the bone marrow and peripheral blood. Although AML can occur at any age, the incidence peaks at age 65. The pathobiology of AML also varies with age with associated differences in incidence, as well as the frequency of cytogenetic change and somatic mutations. In addition, 5-year survival rates in paediatrics are 60-75% but fall to 5-15% in older AML patients. This systematic review aimed to determine whether the altered genes in AML affect the same molecular pathways, indifferent of patient age, and, therefore, whether patients could benefit from the repurposing drugs or the use of the same immunotherapeutic strategies across age boundaries to prevent relapse. Using a PICO framework and PRISMA-P checklist, relevant publications were identified using five literature databases and assessed against an inclusion criteria, leaving 36 articles, and 71 targets for therapy, for further analysis. QUADAS-2 was used to determine the risk of bias and perform a quality control step. We then priority-ranked the list of cancer antigens based on predefined and pre-weighted objective criteria as part of an analytical hierarchy process used for dealing with complex decisions. This organized the antigens according to their potential to act as targets for the immunotherapy of AML, a treatment that offers an opportunity to remove residual leukaemia cells at first remission and improve survival rates. It was found that 80% of the top 20 antigens identified in paediatric AML were also within the 20 highest scoring immunotherapy targets in adult AML. To analyse the relationships between the targets and their link to different molecular pathways, PANTHER and STRING analyses were performed on the 20 highest scoring immunotherapy targets for both adult and paediatric AML. There were many similarities in the PANTHER and STRING results, including the most prominent pathways being angiogenesis and inflammation mediated by chemokine and cytokine signalling pathways. The coincidence of targets suggests that the repurposing of immunotherapy drugs across age boundaries could benefit AML patients, especially when used in combination with conventional therapies. However, due to cost implications, we would recommend that efforts are focused on ways to target the highest scoring antigens, such as WT1, NRAS, IDH1 and TP53, although in the future other candidates may prove successful.
Collapse
Affiliation(s)
- Vanessa S. Morris
- Department of Chemistry and Biochemistry, University of Hull, Kingston upon Hull HU6 7RX, UK;
| | - Hanya Ghazi
- Hull York Medical School, University of Hull, Kingston upon Hull HU6 7RX, UK;
| | - Daniel M. Fletcher
- Centre for Biomedicine, Hull York Medical School, Kingston upon Hull HU6 7RX, UK;
| | - Barbara-ann Guinn
- Centre for Biomedicine, Hull York Medical School, Kingston upon Hull HU6 7RX, UK;
| |
Collapse
|
13
|
Mu X, Chen C, Dong L, Kang Z, Sun Z, Chen X, Zheng J, Zhang Y. Immunotherapy in leukaemia. Acta Biochim Biophys Sin (Shanghai) 2023; 55:974-987. [PMID: 37272727 PMCID: PMC10326417 DOI: 10.3724/abbs.2023101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/19/2023] [Indexed: 06/06/2023] Open
Abstract
Leukaemia is the common name for a group of malignant diseases of the haematopoietic system with complex classifications and characteristics. Remarkable progress has been made in basic research and preclinical studies for acute leukaemia compared to that of the many other types/subtypes of leukaemia, especially the exploration of the biological basis and application of immunotherapy in acute myeloid leukaemia (AML) and B-cell acute lymphoblastic leukaemia (B-ALL). In this review, we summarize the basic approaches to immunotherapy for leukaemia and focus on the research progress made in immunotherapy development for AML and ALL. Importantly, despite the advances made to date, big challenges still exist in the effectiveness of leukaemia immunotherapy, especially in AML. Therefore, we use AML as an example and summarize the mechanisms of tumour cell immune evasion, describe recently reported data and known therapeutic targets, and discuss the obstacles in finding suitable treatment targets and the results obtained in recent clinical trials for several types of single and combination immunotherapies, such as bispecific antibodies, cell therapies (CAR-T-cell treatment), and checkpoint blockade. Finally, we summarize novel immunotherapy strategies for treating lymphocytic leukaemia and clinical trial results.
Collapse
Affiliation(s)
- Xingmei Mu
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chumao Chen
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Loujie Dong
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhaowei Kang
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Zhixian Sun
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Xijie Chen
- Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Junke Zheng
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Yaping Zhang
- Hongqiao International Institute of MedicineShanghai Tongren Hospital/Faculty of Basic MedicineKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
14
|
Zhang W, Wang J, Li W, Liu X, Zhao Y, Yang P, Zhu M, Hu K, Li S, Dong G, Yan C, He X, Zhang X, Jing H. The expression level of Neuronal Calcium Sensor 1 can predict the prognosis of cytogenetically normal AML. THE PHARMACOGENOMICS JOURNAL 2023:10.1038/s41397-023-00301-2. [PMID: 36918700 DOI: 10.1038/s41397-023-00301-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/01/2023] [Accepted: 03/01/2023] [Indexed: 03/15/2023]
Abstract
Acute myeloid leukemia (AML) is malignant clonal expansion of myeloid blasts with high heterogeneity and numerous molecular biomarkers have been found to judge the prognosis in some specific classifications of AML. Furthermore, as for patients with cytogenetically normal acute myeloid leukemia (CN-AML), we need to find more new biomarkers to predict the patients' outcomes. Recently, the expression level of Neuronal Calcium Sensor 1 (NCS1) has been associated with the prognosis of breast cancer and hepatocellular carcinoma, but nothing related has been reported about hematological malignancies. Therefore, we make this study to explore the relationship between the NCS1 expression level and CN-AML. We analyzed the relation between survival and NCS1 RNA expression through 75 CN-AML patients from Cancer Genome Atlas (TCGA) database and 433 CN-AML patients (3 independent datasets) from Gene Expression Omnibus (GEO) database. Additionally, we compared the NCS1 RNA expression between 138 leukemia stem cells positive (LSCs+) samples and 89 leukemia stem cells negative (LSCs-) samples from 78 AML patients from GSE76004 dataset. In our study, CN-AML patients with high expression level of NCS1 have longer EFS or OS. In addition, the NCS1 expression level in leukemia stem cells was low (p = 0.00039). According to these findings, we concluded that the high expression of NCS1 can predict favorable prognosis in CN-AML patients. Furthermore, our work put forward that NCS1 expresses lower in LSCs+, which might be an important mechanism to explain the aggressiveness of AML.
Collapse
Affiliation(s)
- Weilong Zhang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China
| | - Jing Wang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China
| | - Wei Li
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China
| | - Xiaoni Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Yali Zhao
- General Practice Medicine, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Ping Yang
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China
| | - Mingxia Zhu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China
| | - Kai Hu
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China
| | - Shaoxiang Li
- Department of Pathology, Beijing Tiantan Hospital Affiliated with Capital Medical University, 100050, Beijing, China
| | - Gehong Dong
- Department of Pathology, Beijing Tiantan Hospital Affiliated with Capital Medical University, 100050, Beijing, China
| | - Changjian Yan
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China. .,Gannan Medical University, Ganzhou, 341000, China.
| | - Xue He
- Department of Pathology, Beijing Tiantan Hospital Affiliated with Capital Medical University, 100050, Beijing, China.
| | - Xiuru Zhang
- Department of Pathology, Beijing Tiantan Hospital Affiliated with Capital Medical University, 100050, Beijing, China.
| | - Hongmei Jing
- Department of Hematology, Lymphoma Research Center, Peking University Third Hospital, 100191, Beijing, China.
| |
Collapse
|
15
|
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
|
16
|
TP53 Mutant Acute Myeloid Leukemia: The Immune and Metabolic Perspective. HEMATO 2022. [DOI: 10.3390/hemato3040050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TP53 mutated/deleted acute myeloid leukemia (AML) stands out as one of the poorest prognosis forms of acute leukemia with a median overall survival not reaching one year in most cases, even in selected cases when allogenic stem-cell transplantation is performed. This aggressive behavior relies on intrinsic chemoresistance of blast cells and on high rates of relapse. New insights into the biology of the disease have shown strong linkage between TP53 mutant AML, altered metabolic features and immunoregulation uncovering new scenarios and leading to possibilities beyond current treatment approaches. Furthermore, new targeted therapies acting on misfolded/dysfunctional p53 protein are under current investigation with the aim to improve outcomes. In this review, we sought to offer an insight into TP53 mutant AML current biology and treatment approaches, with a special focus on leukemia-associated immune and metabolic changes.
Collapse
|
17
|
Ferguson AL, Sharman AR, Allen RO, Ye T, Lee JH, Low THH, Ch'ng S, Palme CE, Ashford B, Ranson M, Clark JR, Patrick E, Gupta R, Palendira U. High-Dimensional and Spatial Analysis Reveals Immune Landscape-Dependent Progression in Cutaneous Squamous Cell Carcinoma. Clin Cancer Res 2022; 28:4677-4688. [PMID: 36044477 DOI: 10.1158/1078-0432.ccr-22-1332] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/10/2022] [Accepted: 08/29/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE The tumor immune microenvironment impacts the biological behavior of the tumor, but its effect on clinical outcomes in head and neck cutaneous squamous cell carcinomas (HNcSCC) is largely unknown. EXPERIMENTAL DESIGN We compared the immune milieu of high-risk HNcSCC that never progressed to metastasis with those that metastasized using multiparameter imaging mass cytometry. The cohort included both immunosuppressed patients (IS) and patients with an absence of clinical immune-suppression (ACIS). Spatial analyses were used to identify cellular interactions that were associated with tumor behavior. RESULTS Nonprogressing primary HNcSCC were characterized by higher CD8+ and CD4+ T-cell responses, including numerically increased regulatory T cells. In contrast, primary lesions from HNcSCC patients who progressed were largely devoid of T cells with lower numbers of innate immune cells and increased expression of checkpoint receptors and in the metastatic lesions were characterized by an accumulation of B cells. Spatial analysis reveals multiple cellular interactions associated with nonprogressing primary tumors that were distinct in primary tumors of disease-progressing patients. Cellular regional analysis of the tumor microenvironment also shows squamous cell-enriched tumor regions associated with primary nonprogressing tumors. CONCLUSIONS Effective responses from both CD8+ and CD4+ T cells in the tumor microenvironment are essential for immune control of primary HNcSCC. Our findings indicate that the early events that shape the immune responses in primary tumors dictate progression and disease outcomes in HNcSCC.
Collapse
Affiliation(s)
- Angela L Ferguson
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Ashleigh R Sharman
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Ruth O Allen
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Thomas Ye
- School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia
| | - Jenny H Lee
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Tsu-Hui H Low
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,The Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Sydney Ch'ng
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,The Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia.,Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Carsten E Palme
- The Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Bruce Ashford
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia.,School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, New South Wales, Australia
| | - Jonathan R Clark
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,The Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia.,Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, New South Wales, Australia
| | - Ellis Patrick
- School of Mathematics and Statistics, University of Sydney, Sydney, New South Wales, Australia.,Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Ruta Gupta
- Central Clinical School, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW Health Pathology, Sydney, New South Wales, Australia
| | - Umaimainthan Palendira
- Infection, Immunity and Inflammation Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,Centenary Institute, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
18
|
Molica M, Perrone S. Molecular targets for the treatment of AML in the forthcoming 5th World Health Organization Classification of Haematolymphoid Tumours. Expert Rev Hematol 2022; 15:973-986. [PMID: 36271671 DOI: 10.1080/17474086.2022.2140137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a genetically heterogeneous disease for which the treatment armamentarium has been historically restricted to chemotherapy. However, genomic and epigenomic alterations that contribute to AML initiation, maintenance, and relapse have disclosed new insights to the 5th update in WHO Classification of Haematolymphoid Tumours. AREAS COVERED After four decades of intensive chemotherapy as a 'one-size-fits-all' concept, several targeted agents have been approved for the treatment of AML. Several compounds, directed against regulators of apoptotic, epigenetic, or micro-environmental pathways, and immune-system modulators, are currently in development and investigation in clinical trials. We review advances in target-based therapy for AML focusing on their mechanism of action, examining the intracellular events and pathways, and the results from published clinical trials. EXPERT OPINION To improve patient clinical outcomes, find new biomarkers for therapeutic response, and pinpoint patients who might benefit from novel targeted medicines, next-generation sequencing is being used to evaluate AML-associated mutations. In fact, the new 5th edition of WHO classification has reaffirmed the importance of genetically defined entities that have a prognostic impact, but not all have a specific treatment available. New class of target drugs are in clinical development and could be beneficial to improve the therapeutic armamentarium available.
Collapse
Affiliation(s)
| | - Salvatore Perrone
- Hematology, Polo Universitario Pontino, S.M. Goretti Hospital, Latina, Italy.,Division of Hematology, University Hospital Paolo Giaccone, Palermo, Italy
| |
Collapse
|
19
|
Weeda V, Mestrum SGC, Leers MPG. Flow Cytometric Identification of Hematopoietic and Leukemic Blast Cells for Tailored Clinical Follow-Up of Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms231810529. [PMID: 36142442 PMCID: PMC9506284 DOI: 10.3390/ijms231810529] [Citation(s) in RCA: 7] [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: 07/26/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is a myeloid malignancy that is characterized by the accumulation of leukemic blast cells, which originate from hematopoietic stem cells that have undergone leukemic transformation and/or are more mature progenitors that have gained stemness features. Currently, no consensus exists for the flow cytometric identification of normal blast cells and their leukemic counterparts by their antigenic expression profile. Differentiating between the benign cells and the malignant cells is crucial for the further deployment of immunophenotype panels for the clinical follow-up of AML patients. This review provides an overview of immunophenotypic markers that allow the identification of leukemic blast cells in the bone marrow with multiparameter flow cytometry. This technique allows the identification of hematopoietic blast cells at the level of maturing cells by their antigen expression profile. While aberrant antigen expression of a single immunophenotypic marker cell cannot be utilized in order to differentiate leukemic blast cells from normal blast cells, combinations of multiple immunophenotypic markers can enable the distinction of normal and leukemic blast cells. The identification of these markers has provided new perspectives for tailored clinical follow-up, including therapy management, diagnostics, and prognostic purposes. The immunophenotypic marker panels, however, should be developed by carefully considering the variable antigen marker expression profile of individual patients.
Collapse
Affiliation(s)
- Vera Weeda
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
| | - Stefan G. C. Mestrum
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
- Department of Molecular Cell Biology, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre, 6200MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-6-36176124
| | - Math P. G. Leers
- Department of Clinical Chemistry & Hematology, Zuyderland Medical Centre, 6162BG Sittard-Geleen, The Netherlands
| |
Collapse
|
20
|
Lewis AH, Bridges CS, Moorshead DN, Chen TJ, Du W, Zorman B, Sumazin P, Puppi M, Lacorazza HD. Krüppel-like Factor 4 Supports the Expansion of Leukemia Stem Cells in MLL-AF9-driven Acute Myeloid Leukemia. Stem Cells 2022; 40:736-750. [PMID: 35535819 PMCID: PMC9406610 DOI: 10.1093/stmcls/sxac033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 04/27/2022] [Indexed: 11/13/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow with 5-year overall survival of less than 10% in patients over the age of 65. Limited progress has been made in the patient outcome because of the inability to selectively eradicate the leukemic stem cells (LSC) driving the refractory and relapsed disease. Herein, we investigated the role of the reprogramming factor KLF4 in AML because of its critical role in the self-renewal and stemness of embryonic and cancer stem cells. Using a conditional Cre-lox Klf4 deletion system and the MLL-AF9 retroviral mouse model, we demonstrated that loss-of-KLF4 does not significantly affect the induction of leukemia but markedly decreased the frequency of LSCs evaluated in limiting-dose transplantation studies. Loss of KLF4 in leukemic granulocyte-macrophage progenitors (L-GMP), a population enriched for AML LSCs, showed lessened clonogenicity and percentage in the G2/M phase of the cell cycle. RNAseq analysis of purified L-GMPs revealed decreased expression of stemness genes and MLL-target genes and upregulation of the RNA sensing helicase DDX58. However, silencing of DDX58 in KLF4 knockout leukemia indicated that DDX58 is not mediating this phenotype. CRISPR/Cas9 deletion of KLF4 in MOLM13 cell line and AML patient-derived xenograft cells showed impaired expansion in vitro and in vivo associated with a defective G2/M checkpoint. Collectively, our data suggest a mechanism in which KLF4 promotes leukemia progression by establishing a gene expression profile in AML LSCs supporting cell division and stemness.
Collapse
Affiliation(s)
- Andrew Henry Lewis
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - Cory Seth Bridges
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - David Neal Moorshead
- Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Taylor J Chen
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - Wa Du
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - Barry Zorman
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Present address: Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Monica Puppi
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| | - H Daniel Lacorazza
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, TX, USA
| |
Collapse
|
21
|
Zippel S, Dilger N, Chatterjee C, Raic A, Brenner-Weiß G, Schadzek P, Rapp BE, Lee-Thedieck C. A parallelized, perfused 3D triculture model of leukemia for in vitro drug testing of chemotherapeutics. Biofabrication 2022; 14. [PMID: 35472717 DOI: 10.1088/1758-5090/ac6a7e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 04/26/2022] [Indexed: 11/11/2022]
Abstract
Leukemia patients undergo chemotherapy to combat the leukemic cells (LCs) in the bone marrow. During therapy not only the LCs, but also the blood-producing hematopoietic stem and progenitor cells (HSPCs) may be destroyed. Chemotherapeutics targeting only the LCs are urgently needed to overcome this problem and minimize life-threatening side-effects. Predictive in vitro drug testing systems allowing simultaneous comparison of various experimental settings would enhance the efficiency of drug development. Here, we present a 3D human leukemic bone marrow model perfused using a magnetic, parallelized culture system to ensure media exchange. Chemotherapeutic treatment of the acute myeloid leukemia cell line KG-1a in 3D magnetic hydrogels seeded with mesenchymal stem/stromal cells (MSCs) revealed a greater resistance of KG-1a compared to 2D culture. In 3D tricultures with HSPCs, MSCs and KG-1a, imitating leukemic bone marrow, HSPC proliferation decreased while KG-1a cells remained unaffected post treatment. Non-invasive metabolic profiling enabled continuous monitoring of the system. Our results highlight the importance of using biomimetic 3D platforms with proper media exchange and co-cultures for creating in vivo-like conditions to enable in vitro drug testing. This system is a step towards drug testing in biomimetic, parallelized in vitro approaches, facilitating the discovery of new anti-leukemic drugs.
Collapse
Affiliation(s)
- Sabrina Zippel
- Institute of Cell Biology and Biophysics, Leibniz Universitat Hannover, Herrenhäuser Str. 2, Hannover, 30419, GERMANY
| | - Nadine Dilger
- Institute of Cell Biology and Biophysics, Leibniz University Hanover, Herrenhäuser Str. 2, Hannover, 30419, GERMANY
| | - Chandralekha Chatterjee
- Institute of Cell Biology and Biophysics, Leibniz Universitat Hannover, Herrenhäuser Str. 2, Hannover, 30419, GERMANY
| | - Annamarija Raic
- Institute of Cell Biology and Biophysics, Leibniz Universitat Hannover, Herrenhäuser Str. 2, Hannover, 30419, GERMANY
| | - Gerald Brenner-Weiß
- Institute of Functional Interfaces, Karlsruhe Institute of Technology, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Baden-Württemberg, 76344, GERMANY
| | - Patrik Schadzek
- Department of Orthopedic Surgery, Graded Implants and Regenerative Strategies, OE 8893, Laboratory for Biomechanics and Biomaterials, Hannover Medical School, Stadtfelddamm 34, Hannover, Niedersachsen, 30625, GERMANY
| | - Bastian E Rapp
- Department of Microsystems Engineering (IMTEK), Albert-Ludwigs-Universitat Freiburg, Georges-Köhler-Allee 103, Freiburg im Breisgau, Baden-Württemberg, 79110, GERMANY
| | - Cornelia Lee-Thedieck
- Institute of Cell Biology and Biophysics, Leibniz Universitat Hannover, Herrenhäuser Str. 2, Hannover, 30419, GERMANY
| |
Collapse
|
22
|
Hong J, Xia L, Huang Z, Yuan X, Liang X, Dai J, Wu Z, Liang L, Ruan M, Long Z, Cheng X, Chen X, Ni J, Ge J, Li Q, Zeng Q, Xia R, Wang Y, Yang M. TIM-3 Expression Level on AML Blasts Correlates With Presence of Core Binding Factor Translocations Rather Than Clinical Outcomes. Front Oncol 2022; 12:879471. [PMID: 35494006 PMCID: PMC9046698 DOI: 10.3389/fonc.2022.879471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background T-cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3) expresses on leukemic stem and progenitor populations of non-M3 acute myeloid leukemia (AML) as well as T lymphocytes. TIM-3 is thought to be involved in the self-renewal of leukemic stem cells and the immune escape of AML cells, however its correlation with AML prognosis is still controversial and worthy of further investigation. Methods we simultaneously assessed TIM-3 expression levels of leukemic blasts and T lymphocytes in the bone marrow of de novo AML patients using flow cytometry. The correlations of TIM-3 expression between leukemic blasts and T lymphocytes and the correlations of TIM-3 expression with various patient parameters were analyzed. In addition, the Cancer Genome Atlas (TCGA) data of AML patients were acquired and analyzed to verify the results. Results TIM-3 expression of CD34+ leukemic blasts (R2 = 0.95, p<0.0001) and CD34+CD38- leukemic stem cells (R2 = 0.75, p<0.0001) were significantly and positively correlated with that of the whole population of leukemic blasts. In addition, TIM-3 expression level of leukemic blasts correlated significantly and positively with that of CD8+ (R2 = 0.44, p<0.0001) and CD4+ (R2 = 0.16, p=0.0181) lymphocytes, and higher TIM-3 expression of leukemic blasts was significantly associated with a greater proportion of peripheral CD8+ T lymphocytes (R2 = 0.24, p=0.0092), indicating that TIM-3 on leukemic blasts might alter adaptive immunity of AML patients. Regarding clinical data, the presence of core binding factor (CBF) translocations was significantly correlated with higher TIM-3 expression of leukemic blasts (CBF versus non-CBF, median 22.78% versus 1.28%, p=0.0012), while TIM-3 expression levels of leukemic blasts were not significantly associated with the remission status after induction chemotherapy (p=0.9799), overall survival (p=0.4201) or event-free survival (p=0.9873). Similar to our results, TCGA data showed that patients with CBF translocations had significantly higher mRNA expression level of HAVCR2 (the gene encoding TIM-3) (median, 9.81 versus 8.69, p<0.0001), and as all patients in the cohort were divided into two groups based on the median HAVCR2 expression level, 5-year overall survivals were not significantly different (low versus high, 24.95% versus 24.54%, p=0.6660). Conclusion TIM-3 expression level on AML blasts correlates with presence of CBF translocations rather than clinical outcomes.
Collapse
Affiliation(s)
- Jian Hong
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Leiming Xia
- Department of Hematology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhenqi Huang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaodong Yuan
- Division of Life Sciences and Medicine, Department of Organ Transplantation Center, Transplant and Immunology Laboratory, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
| | - Xinglin Liang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jifei Dai
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhonghui Wu
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Liang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Ruan
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhangbiao Long
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Cheng
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaowen Chen
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Ni
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jian Ge
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qingsheng Li
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qingshu Zeng
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruixiang Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Wang
- Department of Oncology, The Third Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Mingzhen Yang, ; Yi Wang,
| | - Mingzhen Yang
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Hematology, The Forth Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Mingzhen Yang, ; Yi Wang,
| |
Collapse
|
23
|
Hidalgo I, Wahlestedt M, Yuan O, Zhang Q, Bryder D, Pronk CJ. Bmi1 induction protects hematopoietic stem cells against pronounced long-term hematopoietic stress. Exp Hematol 2022; 109:35-44. [DOI: 10.1016/j.exphem.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 11/04/2022]
|
24
|
Ayyadurai VAS, Deonikar P, McLure KG, Sakamoto KM. Molecular Systems Architecture of Interactome in the Acute Myeloid Leukemia Microenvironment. Cancers (Basel) 2022; 14:756. [PMID: 35159023 PMCID: PMC8833542 DOI: 10.3390/cancers14030756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/29/2022] [Indexed: 12/12/2022] Open
Abstract
A molecular systems architecture is presented for acute myeloid leukemia (AML) to provide a framework for organizing the complexity of biomolecular interactions. AML is a multifactorial disease resulting from impaired differentiation and increased proliferation of hematopoietic precursor cells involving genetic mutations, signaling pathways related to the cancer cell genetics, and molecular interactions between the cancer cell and the tumor microenvironment, including endothelial cells, fibroblasts, myeloid-derived suppressor cells, bone marrow stromal cells, and immune cells (e.g., T-regs, T-helper 1 cells, T-helper 17 cells, T-effector cells, natural killer cells, and dendritic cells). This molecular systems architecture provides a layered understanding of intra- and inter-cellular interactions in the AML cancer cell and the cells in the stromal microenvironment. The molecular systems architecture may be utilized for target identification and the discovery of single and combination therapeutics and strategies to treat AML.
Collapse
Affiliation(s)
- V. A. Shiva Ayyadurai
- Systems Biology Group, International Center for Integrative Systems, Cambridge, MA 02138, USA;
| | - Prabhakar Deonikar
- Systems Biology Group, International Center for Integrative Systems, Cambridge, MA 02138, USA;
| | | | - Kathleen M. Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA;
| |
Collapse
|
25
|
Tettamanti S, Pievani A, Biondi A, Dotti G, Serafini M. Catch me if you can: how AML and its niche escape immunotherapy. Leukemia 2022; 36:13-22. [PMID: 34302116 PMCID: PMC8727297 DOI: 10.1038/s41375-021-01350-x] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023]
Abstract
In spite of the remarkable progress in basic and preclinical studies of acute myeloid leukemia (AML), the five-year survival rate of AML patients remains poor, highlighting the urgent need for novel and synergistic therapies. Over the past decade, increased attention has been focused on identifying suitable immunotherapeutic strategies for AML, and in particular on targeting leukemic cells and their progenitors. However, recent studies have also underlined the important contribution of the leukemic microenvironment in facilitating tumor escape mechanisms leading to disease recurrence. Here, we describe the immunological features of the AML niche, with particular attention to the crosstalk between the AML blasts and the cellular components of the altered tumor microenvironment (TME) and the mechanisms of immune escape that hamper the therapeutic effects of the most advanced treatments. Considering the AML complexity, immunotherapy approaches may benefit from a rational combination of complementary strategies aimed at preventing escape mechanisms without increasing toxicity.
Collapse
Affiliation(s)
- Sarah Tettamanti
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Alice Pievani
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| | - Andrea Biondi
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy.
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marta Serafini
- Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca/Fondazione MBBM, Monza, Italy
| |
Collapse
|
26
|
Rezaei M, Tan J, Zeng C, Li Y, Ganjalikhani-Hakemi M. TIM-3 in Leukemia; Immune Response and Beyond. Front Oncol 2021; 11:753677. [PMID: 34660319 PMCID: PMC8514831 DOI: 10.3389/fonc.2021.753677] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/10/2021] [Indexed: 02/05/2023] Open
Abstract
T cell immunoglobulin and mucin domain 3 (TIM-3) expression on malignant cells has been reported in some leukemias. In myelodysplastic syndrome (MDS), increased TIM-3 expression on TH1 cells, regulatory T cells, CD8+ T cells, and hematopoietic stem cells (HSCs), which play a role in the proliferation of blasts and induction of immune escape, has been reported. In AML, several studies have reported overexpression of TIM-3 on leukemia stem cells (LSCs) but not on healthy HSCs. Overexpression of TIM-3 on exhausted CD4+ and CD8+ T cells and leukemic cells in CML, ALL, and CLL patients could be a prognostic risk factor for poor therapeutic response and relapse in patients. Currently, several TIM-3 inhibitors are used in clinical trials for leukemias, and some have shown encouraging response rates for MDS and AML treatment. For AML immunotherapy, blockade TIM-3 may have dual effects: directly inhibiting AML cell proliferation and restoring T cell function. However, blockade of PD-1 and TIM-3 fails to restore the function of exhausted CD8+ T cells in the early clinical stages of CLL, indicating that the effects of TIM-3 blockade may be different in AML and other leukemias. Thus, further studies are required to evaluate the efficacy of TIM-3 inhibitors in different types and stages of leukemia. In this review, we summarize the biological functions of TIM-3 and its contribution as it relates to leukemias. We also discuss the effects of TIM-3 blockade in hematological malignancies and clinical trials of TIM-3 for leukemia therapy.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jiaxiong Tan
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chengwu Zeng
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Mazdak Ganjalikhani-Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Acquired Immunodeficiency Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
27
|
Chajuwan T, Kansuwan P, Kobbuaklee S, Chanswangphuwana C. Characteristics and clinical correlation of TIM-3 and PD-1/PD-L1 expressions in leukemic cells and tumor microenvironment in newly diagnosed acute myeloid leukemia. Leuk Lymphoma 2021; 63:450-456. [PMID: 34585994 DOI: 10.1080/10428194.2021.1984454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Dual targeting of TIM-3 and PD-1/PD-L1 pathways is currently under investigation for cancer immunotherapy. The interaction of these immune checkpoints remains unclear in the leukemic microenvironment of acute myeloid leukemia (AML). We performed an immunophenotypic study of bone marrow in 37 newly diagnosed AML patients. High levels of TIM-3 expression on AML blasts were correlated with first 7 + 3 induction failure (CR 16.2% vs. non-CR 36.4%, p = .038). In contrast, high TIM-3 levels on natural killer (NK) cells were associated with complete remission (CR) status after induction (CR 24.7% vs. non-CR 6.5%, p = .035). Few PD-L1 positive AML blasts and PD-1 or PD-L1 positive NK cells were observed. Although the exhausted PD-1 expressing T cells were detected in 28.3% of T cells, the double positive of PD-1 and TIM-3 T cells were rarely detected. In summary, the TIM-3 levels on AML blasts and NK cells are potentially the prognostic biomarkers in AML.
Collapse
Affiliation(s)
- Thunyamon Chajuwan
- Department of Medicine, Division of Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand.,Research Unit in Translational Hematology, Chulalongkorn University, Bangkok, Thailand
| | - Patsita Kansuwan
- Department of Medicine, Division of Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Sirorat Kobbuaklee
- Department of Medicine, Division of Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chantiya Chanswangphuwana
- Department of Medicine, Division of Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand.,Research Unit in Translational Hematology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
28
|
Wang J, Yang L, Dao FT, Wang YZ, Chang Y, Xu N, Chen WM, Jiang Q, Jiang H, Liu YR, Qin YZ. Prognostic significance of TIM-3 expression pattern at diagnosis in patients with t(8;21) acute myeloid leukemia. Leuk Lymphoma 2021; 63:152-161. [PMID: 34405769 DOI: 10.1080/10428194.2021.1966785] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Acute myeloid leukemia (AML) with t(8;21) is a heterogeneous disease and needs to be stratified. Both, cancer cells and immune cells participate in tumor initiation, growth and progression and might affect clinical outcomes. TIM-3 (T cell immunoglobulin and mucin domain-containing protein 3), an immune checkpoint molecule, is expressed not only on immune cells but also on leukemic stem cells (LSCs) in AML. This prompted us to investigate the prognostic significance of TIM-3 in t(8;21) AML. A total of 47 t(8;21) AML patients were tested for TIM-3 expression by multi-parameter flow cytometry at diagnosis. 35 of these, who received chemotherapy alone or along with allogeneic hematopoietic stem cell transplantation were followed up. The expression pattern of TIM-3 on T-cells and NK (natural killer) cells as a whole (T + NK) and LSCs were evaluated independently. High percentage of T + NK - TIM-3+ and CD34+CD38-TIM-3+ cells were significantly associated with a high 2-year cumulative incidence of relapse (CIR) (p = 0.028, 0.016). Further, concurrent high frequencies of T + NK-TIM-3+ and CD34+CD38-TIM-3+ cells at diagnosis were significantly associated with a high 2-year CIR (p < 0.0001) and this together with c-KIT D816 mutation were the independent adverse prognostic factors for relapse (hazard ratio (HR)=2.5, [95% confidence interval (CI), 1.1-6.0], p = 0.04; HR = 46.5, [95% CI, 2.7-811.5], p = 0.009). In conclusion, the expression pattern of TIM-3 on both T and NK cells and LSCs at diagnosis had prognostic significance in t (8;21) AML.
Collapse
Affiliation(s)
- Jun Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Lu Yang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Feng-Ting Dao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Ya-Zhe Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yan Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Nan Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Wen-Min Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Qian Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Hao Jiang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Yan-Rong Liu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| | - Ya-Zhen Qin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing, China
| |
Collapse
|
29
|
Sudha T, Godugu K, Darwish NHE, Nazeer T, Mousa SA. Novel Polyethylene Glycol-Conjugated Triazole Derivative with High Thyrointegrin αvβ3 Affinity in Acute Myeloid Leukemia Management. Cancers (Basel) 2021; 13:cancers13164070. [PMID: 34439224 PMCID: PMC8392871 DOI: 10.3390/cancers13164070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023] Open
Abstract
(1) Background: Acute myeloid leukemia (AML) accounts for up to one-third of more than 60,000 leukemia cases diagnosed annually in the U.S. Primary AML cells express membrane αvβ3 integrin, which is associated with adverse prognosis and resistance to chemotherapies. A novel anticancer compound Polyethylene glycol-conjugated bi-TriAzole Tetraiodothyroacetic acid (P-bi-TAT) interacts with high affinity (Ki 0.3 nM) and specificity with the thyrointegrin αvβ3. We evaluated P-bi-TAT activities in two different AML models representing monocytic and myelocytic forms of acute leukemia. (2) Methods and Results: The in vivo AML models were established prior to initiation of treatment protocols by grafting human leukemia cells in immunocompromised mice. IVIS imaging scans revealed that leukemic colonies were extensively established throughout the bone marrow, liver, and lung of the untreated animals. In animals treated with P-bi-TAT at daily doses ranging from 1-10 mg/kg, subcutaneously for 2-3 weeks, IVIS imaging scans revealed 95% reduction in bone marrow colonies and leukemic colonies in liver and lung. Also, the leukemic cells were not detected in bone marrow samples of P-bi-TAT-treated animals. The anti-neoplastic effect of P-bi-TAT administration on leukemic cells was associated with marked inhibition of NF-κB activity. We conclude that experimental P-bi-TAT therapy in vivo appears extraordinarily effective against the two forms of human AML models in mice. Because the P-bi-TAT molecular target, thyrointegrin αvβ3, is consistently expressed in many, if not all, clinical AML samples, P-bi-TAT-based therapy seems to have significant clinical potential in treating most AML sub-types. Hence, P-bi-TAT represents a promising targeted therapeutic agent for AML patients.
Collapse
Affiliation(s)
- Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
| | - Kavitha Godugu
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
| | - Noureldien H. E. Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
- Hematology Unit, Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Tipu Nazeer
- Albany Medical Center, Pathology Department, AMC Hospital, Albany, NY 12208, USA;
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA; (T.S.); (K.G.); (N.H.E.D.)
- Correspondence:
| |
Collapse
|
30
|
Depreter B, De Moerloose B, Vandepoele K, Uyttebroeck A, Van Damme A, Terras E, Denys B, Dedeken L, Dresse MF, Van der Werff Ten Bosch J, Hofmans M, Philippé J, Lammens T. Deciphering molecular heterogeneity in pediatric AML using a cancer vs. normal transcriptomic approach. Pediatr Res 2021; 89:1695-1705. [PMID: 33069162 DOI: 10.1038/s41390-020-01199-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/30/2020] [Accepted: 09/25/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Still 30-40% of pediatric acute myeloid leukemia (pedAML) patients relapse. Delineation of the transcriptomic profile of leukemic subpopulations could aid in a better understanding of molecular biology and provide novel biomarkers. METHODS Using microarray profiling and quantitative PCR validation, transcript expression was measured in leukemic stem cells (LSC, n = 24) and leukemic blasts (L-blast, n = 25) from pedAML patients in comparison to hematopoietic stem cells (HSCs, n = 19) and control myeloblasts (C-blast, n = 20) sorted from healthy subjects. Gene set enrichment analysis was performed to identify relevant gene set enrichment signatures, and functional protein associations were identified by STRING analysis. RESULTS Highly significantly overexpressed genes in LSC and L-blast were identified with a vast majority not studied in AML. CDKN1A, CFP, and CFD (LSC) and HOMER3, CTSA, and GADD45B (L-blast) represent potentially interesting biomarkers and therapeutic targets. Eleven LSC downregulated targets were identified that potentially qualify as tumor suppressor genes, with MYCT1, PBX1, and PTPRD of highest interest. Inflammatory and immune dysregulation appeared to be perturbed biological networks in LSC, whereas dysregulated metabolic profiles were observed in L-blast. CONCLUSION Our study illustrates the power of taking into account cell population heterogeneity and reveals novel targets eligible for functional evaluation and therapy in pedAML. IMPACT Novel transcriptional targets were discovered showing a significant differential expression in LSCs and blasts from pedAML patients compared to their normal counterparts from healthy controls. Deregulated pathways, including immune and metabolic dysregulation, were addressed for the first time in children, offering a deeper understanding of the molecular pathogenesis. These novel targets have the potential of acting as biomarkers for risk stratification, follow-up, and targeted therapy. Multiple LSC-downregulated targets endow tumor suppressor roles in other cancer entities, and further investigation whether hypomethylating therapy could result into LSC eradication in pedAML is warranted.
Collapse
Affiliation(s)
- Barbara Depreter
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.
| | - Barbara De Moerloose
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Karl Vandepoele
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Anne Uyttebroeck
- Department of Pediatrics, University Hospital Gasthuisberg, Leuven, Belgium
| | - An Van Damme
- Department of Pediatric Hematology Oncology, University Hospital Saint-Luc, Brussels, Belgium
| | - Eva Terras
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| | - Barbara Denys
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Laurence Dedeken
- Department of Pediatric Hematology Oncology, Queen Fabiola Children's University Hospital, Brussels, Belgium
| | | | | | - Mattias Hofmans
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Jan Philippé
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tim Lammens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
31
|
Kamal AM, Nabih NA, Elleboudy NS, Radwan SM. Expression of immune check point gene TIM-3 in patients newly diagnosed with acute myeloid leukemia: Significance and impact on outcome. Oncol Lett 2021; 21:325. [PMID: 33692857 PMCID: PMC7933755 DOI: 10.3892/ol.2021.12587] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/12/2021] [Indexed: 12/16/2022] Open
Abstract
Despite recent advancements in the therapeutic landscape of acute myeloid leukemia (AML), the prognosis of patients remains poor. Immune check point inhibitors have been investigated in hematological malignancies, including AML; however, the role of T-cell immunoglobulin and mucin domain 3 (TIM-3) in AML has not yet been fully elucidated. Thus, the present study aimed to investigate TIM-3 gene expression in patients with AML and determine its associations with prognostic variables and clinical outcome. A total of 60 patients newly diagnosed with AML and 15 healthy matching individuals were recruited in the present study, and reverse transcription-quantitative PCR analysis was performed to detect TIM-3 expression. The results demonstrated that TIM-3 expression was significantly upregulated in patients with AML compared with that in healthy individuals (P<0.001). In addition, patients with extramedullary disease (EMD) exhibited significantly lower median TIM-3 expression levels compared with those without EMD (P=0.001). Furthermore, patients with high TIM-3 expression had significantly lower complete remission rates following induction chemotherapy compared with those with low TIM-3 expression (P=0.004). High TIM-3 expression was significantly associated with lower overall survival rates during the 1-year follow-up (P=0.001). Taken together, the results of the present study suggest that TIM-3 may act as a biomarker of a poor prognosis in patients with AML, and be used as a therapeutic target.
Collapse
Affiliation(s)
- Amany M. Kamal
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Nermeen A. Nabih
- Department of Internal Medicine, Clinical Hematology and Bone Marrow Transplantation Unit, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt
| | - Nooran S. Elleboudy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Sara M. Radwan
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
32
|
Swatler J, Turos-Korgul L, Kozlowska E, Piwocka K. Immunosuppressive Cell Subsets and Factors in Myeloid Leukemias. Cancers (Basel) 2021; 13:cancers13061203. [PMID: 33801964 PMCID: PMC7998753 DOI: 10.3390/cancers13061203] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Effector immune system cells have the ability to kill tumor cells. However, as a cancer (such as leukemia) develops, it inhibits and evades the effector immune response. Such a state of immunosuppression can be driven by several factors – receptors, soluble cytokines, as well as by suppressive immune cells. In this review, we describe factors and cells that constitute immunosuppressive microenvironment of myeloid leukemias. We characterize factors of direct leukemic origin, such as inhibitory receptors, enzymes and extracellular vesicles. Furthermore, we describe suppressive immune cells, such as myeloid derived suppressor cells and regulatory T cells. Finally, we sum up changes in these drivers of immune evasion in myeloid leukemias during therapy. Abstract Both chronic myeloid leukemia and acute myeloid leukemia evade the immune response during their development and disease progression. As myeloid leukemia cells modify their bone marrow microenvironment, they lead to dysfunction of cytotoxic cells, such as CD8+ T cells or NK cells, simultaneously promoting development of immunosuppressive regulatory T cells and suppressive myeloid cells. This facilitates disease progression, spreading of leukemic blasts outside the bone marrow niche and therapy resistance. The following review focuses on main immunosuppressive features of myeloid leukemias. Firstly, factors derived directly from leukemic cells – inhibitory receptors, soluble factors and extracellular vesicles, are described. Further, we outline function, properties and origin of main immunosuppressive cells - regulatory T cells, myeloid derived suppressor cells and macrophages. Finally, we analyze interplay between recovery of effector immunity and therapeutic modalities, such as tyrosine kinase inhibitors and chemotherapy.
Collapse
Affiliation(s)
- Julian Swatler
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
| | - Laura Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
| | - Ewa Kozlowska
- Department of Immunology, Institute of Functional Biology and Ecology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland; (J.S.); (L.T.-K.)
- Correspondence:
| |
Collapse
|
33
|
Lin G, Zhang Y, Yu L, Wu D. Cytotoxic effect of CLL‑1 CAR‑T cell immunotherapy with PD‑1 silencing on relapsed/refractory acute myeloid leukemia. Mol Med Rep 2021; 23:208. [PMID: 33495835 PMCID: PMC7830996 DOI: 10.3892/mmr.2021.11847] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 11/04/2020] [Indexed: 01/29/2023] Open
Abstract
The activation of chimeric antigen receptor (CAR)-T cells can lead to persistently high levels of programmed cell death 1 (PD-1) antigen and eventually causes the exhaustion of T cells. The effectiveness of CAR-T cells targeting C-type lectin-like molecule-1 (CLL-1) combined with PD-1 silencing therapy for acute myeloid leukemia (AML) was evaluated in the present study. CLL-1 levels in primary AML bone marrow samples was examined using flow cytometric analysis. We designed a CLL-1 CAR-T, containing CLL-1-specific single-chain variable fragment, CD28, OX40, CD8 hinge and TM and CD3-ζ signaling domains. CLL-1 CAR-T with PD-1 silencing was constructed. It was confirmed that CLL-1 is expressed on the surface of AML cells. CLL-1 CAR-T showed specific lysing activity against CLL-1+ AML cells. PD-1 silencing enhanced the killing ability of CLL-1 CAR-T. Furthermore, it was found that CAR-T derived from healthy donor T cells was more effective in killing THP-1 cells (a human acute monocytic leukemia cell line) than those from patient-derived T cells. These results indicated that CLL-1 CAR-T and PD-1 knockdown CLL-1 CAR-T could be used as a potential immunotherapy to treat relapsed or refractory AML.
Collapse
Affiliation(s)
- Guoqiang Lin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yanming Zhang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical College, Huai'an Second People's Hospital, Huai'an, Jiangsu 223002, P.R. China
| | - Lei Yu
- Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200065, P.R. China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
34
|
Taghiloo S, Asgarian-Omran H. Immune evasion mechanisms in acute myeloid leukemia: A focus on immune checkpoint pathways. Crit Rev Oncol Hematol 2020; 157:103164. [PMID: 33271388 DOI: 10.1016/j.critrevonc.2020.103164] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/09/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Immune surveillance mechanisms comprising of adaptive and innate immune systems are naturally designed to eliminate AML development. However, leukemic cells apply various immune evasion mechanisms to deviate host immune responses resulting tumor progression. One of the recently well-known immune escape mechanisms is over-expression of immune checkpoint receptors and their ligands. Introduction of blocking antibodies targeting co-inhibitory molecules achieved invaluable success in tumor targeted therapy. Moreover, several new co-inhibitory pathways are currently studying for their potential impacts on improving anti-tumor immune responses. Although immunotherapeutic strategies based on the blockade of immune checkpoint molecules have shown promising results in a number of hematological malignances, their effectiveness in AML patients showed less remarkable success. This review discusses current knowledge about the involvement of co-inhibitory signaling pathways in immune evasion mechanisms of AML and potential application of immune checkpoint inhibitors for targeted immunotherapy of this malignancy.
Collapse
Affiliation(s)
- Saeid Taghiloo
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
35
|
Ackun-Farmmer MA, Alatise KL, Cross G, Benoit DSW. Ligand Density Controls C-Type Lectin-Like Molecule-1 Receptor-Specific Uptake of Polymer Nanoparticles. ADVANCED BIOSYSTEMS 2020; 4:e2000172. [PMID: 33073549 PMCID: PMC7959326 DOI: 10.1002/adbi.202000172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/01/2020] [Indexed: 01/13/2023]
Abstract
The newest generation of drug delivery systems (DDSs) exploits ligands to mediate specific targeting of cells and/or tissues. However, studies investigating the link between ligand density and nanoparticle (NP) uptake are limited to a small number of ligand-receptor systems. C-type lectin-like molecule-1 (CLL1) is uniquely expressed on myeloid cells, which enables the development of receptors specifically targeting treat various diseases. This study aims to investigate how NPs with different CLL1 targeting peptide density impact cellular uptake. To this end, poly(styrene-alt-maleic anhydride)-b-poly(styrene) NPs are functionalized with cyclized CLL1 binding peptides (cCBP) ranging from 240 ± 12 to 31 000 ± 940 peptides per NP. Unexpectedly, the percentage of cells with internalized NPs is decreased for all cCBP-NP designs regardless of ligand density compared to unmodified NPs. Internalization through CLL1 receptor-mediated processes is further investigated without confounding the effects of NP size and surface charge. Interestingly, high density cCBP-NPs (>7000 cCBP per NP) uptake is dominated by CLL1 receptor-mediated processes while low density cCBP-NPs (≈200 cCBP per NP) and untargeted NP occurred through non-specific clathrin and caveolin-mediated endocytosis. Altogether, these studies show that ligand density and uptake mechanism should be carefully investigated for specific ligand-receptor systems for the design of targeted DDSs to achieve effective drug delivery.
Collapse
Affiliation(s)
- Marian A Ackun-Farmmer
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, USA
- University of Rochester Medical Center, Department of Orthopaedics and Center for Musculoskeletal Research, Rochester, NY, USA
| | - Kharimat L Alatise
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, USA
| | - Griffin Cross
- Washington University in St. Louis, Biomedical/Medical Engineering, St. Louis, MO, USA
| | - Danielle S W Benoit
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, USA
- University of Rochester Medical Center, Department of Orthopaedics and Center for Musculoskeletal Research, Rochester, NY, USA
- University of Rochester, Materials Science Program, Rochester, NY, USA
- University of Rochester, Department of Chemical Engineering, Rochester, NY, USA
| |
Collapse
|
36
|
Tusa I, Cheloni G, Poteti M, Silvano A, Tubita A, Lombardi Z, Gozzini A, Caporale R, Scappini B, Dello Sbarba P, Rovida E. In Vitro Comparison of the Effects of Imatinib and Ponatinib on Chronic Myeloid Leukemia Progenitor/Stem Cell Features. Target Oncol 2020; 15:659-671. [PMID: 32780298 PMCID: PMC7568716 DOI: 10.1007/s11523-020-00741-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background The development of molecularly tailored therapeutic agents such as the BCR/ABL-active tyrosine kinase inhibitors (TKi) resulted in an excellent treatment option for chronic myeloid leukemia (CML) patients. However, following TKi discontinuation, disease relapses in 40–60% of patients, an occurrence very likely due to the persistence of leukemic stem cells that are scarcely sensitive to TKi. Nevertheless, TKi are still the only current treatment option for CML patients. Objective The aim of this study was to compare the effects of TKi belonging to different generations, imatinib and ponatinib (first and third generation, respectively), on progenitor/stem cell expansion potential and markers. Patients and Methods We used stabilized CML cell lines (KCL22, K562 and LAMA-84 cells), taking advantage of the previous demonstration of ours that cell lines contain cell subsets endowed with progenitor/stem cell properties. Primary cells explanted from CML patients were also used. The effects of TKi on the expression of stem cell related genes were compared by quantitative PCR. Flow cytometry was performed to evaluate aldehyde-dehydrogenase (ALDH) activity and the expression of cluster of differentiation (CD) cell surface hematopoietic stem cell markers. Progenitor/stem cell potential was estimated by serial colony formation ability (CFA) assay. Results Ponatinib was more effective than imatinib for the reduction of cells with ALDH activity and progenitor/stem cell potential of CML patient-derived cells and cell lines. Furthermore, ponatinib was more effective than imatinib in reducing the percentage of CD26-expressing cells in primary CML cells, whereas imatinib and ponatinib showed similar efficacy on KCL22 cells. Both drugs strongly upregulated NANOG and SOX2 in CML cell lines, but in KCL22 cells this upregulation was significantly lower with ponatinib than with imatinib, an outcome compatible with a lower level of enrichment of the stem cell compartment upon ponatinib treatment. Conclusion Ponatinib seems to target CML progenitor/stem cells better than imatinib. Electronic supplementary material The online version of this article (10.1007/s11523-020-00741-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Giulia Cheloni
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Martina Poteti
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Angela Silvano
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | - Zoe Lombardi
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy
| | | | - Roberto Caporale
- Dipartimento DAI Oncologico e di Chirurgia ad Indirizzo Robotico SOD Centro Diagnostico di Citofluorimetria e Immunoterapia, AOU Careggi, Florence, Italy
| | | | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Science, University of Florence, Viale GB Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
37
|
Walcher L, Kistenmacher AK, Suo H, Kitte R, Dluczek S, Strauß A, Blaudszun AR, Yevsa T, Fricke S, Kossatz-Boehlert U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front Immunol 2020; 11:1280. [PMID: 32849491 PMCID: PMC7426526 DOI: 10.3389/fimmu.2020.01280] [Citation(s) in RCA: 541] [Impact Index Per Article: 108.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
The use of biomarkers in diagnosis, therapy and prognosis has gained increasing interest over the last decades. In particular, the analysis of biomarkers in cancer patients within the pre- and post-therapeutic period is required to identify several types of cells, which carry a risk for a disease progression and subsequent post-therapeutic relapse. Cancer stem cells (CSCs) are a subpopulation of tumor cells that can drive tumor initiation and can cause relapses. At the time point of tumor initiation, CSCs originate from either differentiated cells or adult tissue resident stem cells. Due to their importance, several biomarkers that characterize CSCs have been identified and correlated to diagnosis, therapy and prognosis. However, CSCs have been shown to display a high plasticity, which changes their phenotypic and functional appearance. Such changes are induced by chemo- and radiotherapeutics as well as senescent tumor cells, which cause alterations in the tumor microenvironment. Induction of senescence causes tumor shrinkage by modulating an anti-tumorigenic environment in which tumor cells undergo growth arrest and immune cells are attracted. Besides these positive effects after therapy, senescence can also have negative effects displayed post-therapeutically. These unfavorable effects can directly promote cancer stemness by increasing CSC plasticity phenotypes, by activating stemness pathways in non-CSCs, as well as by promoting senescence escape and subsequent activation of stemness pathways. At the end, all these effects can lead to tumor relapse and metastasis. This review provides an overview of the most frequently used CSC markers and their implementation as biomarkers by focussing on deadliest solid (lung, stomach, liver, breast and colorectal cancers) and hematological (acute myeloid leukemia, chronic myeloid leukemia) cancers. Furthermore, it gives examples on how the CSC markers might be influenced by therapeutics, such as chemo- and radiotherapy, and the tumor microenvironment. It points out, that it is crucial to identify and monitor residual CSCs, senescent tumor cells, and the pro-tumorigenic senescence-associated secretory phenotype in a therapy follow-up using specific biomarkers. As a future perspective, a targeted immune-mediated strategy using chimeric antigen receptor based approaches for the removal of remaining chemotherapy-resistant cells as well as CSCs in a personalized therapeutic approach are discussed.
Collapse
Affiliation(s)
- Lia Walcher
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ann-Kathrin Kistenmacher
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Reni Kitte
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sarah Dluczek
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Alexander Strauß
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - André-René Blaudszun
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Stephan Fricke
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Uta Kossatz-Boehlert
- Department of Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
38
|
Chen S, Gan S, Han L, Li X, Xie X, Zou D, Sun H. Artesunate induces apoptosis and inhibits the proliferation, stemness, and tumorigenesis of leukemia. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:767. [PMID: 32647692 PMCID: PMC7333094 DOI: 10.21037/atm-20-4558] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Leukemia is characterized by the presence of highly malignant tumors formed in the hematopoietic system. Artesunate (Art), a semi-synthetic derivative of artemisinin, is commonly used as an antimalarial drug and has been proven to possess anticancer potential. Methods In this study, the effect of Art on the proliferation and stemness of human acute promyelocyte leukemia HL-60 cells and acute myeloid leukemia KG1a cells was investigated. Flow cytometry, colony formation assay, the protein expressive levels of survivin, P21, cleaved caspase 3, Bax, Bcl-2, Ki67 were detected the effect of Art on HL-60 and KG1a cells proliferation and apoptosis. At the same time, cell sphere formation assay and the protein expressive levels of CD44, SOX2, ALDH1 and OCT4 were used to analyze the effects of Art on cancer stem cell-like property in vitro. The orthotopic xenograft mouse models were established by using KG1a cells in BALB/c athymic nude mice. Tumor weigh was detected. The protein levels of survivin and Ki67 were detected by immunohistochemistry assays. Results Art induced cell apoptosis and inhibited cell proliferation and stemness in a dose-dependent manner. In the meantime, the results exhibited that Art inhibited the growth and stemness of transplanted tumors via the suppression of the MEK/ERK and PI3K/Akt pathway. Conclusions Our present study provides new insights into the mechanisms of Art’s anticancer potential in leukemia.
Collapse
Affiliation(s)
- Shengmei Chen
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Silin Gan
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lijie Han
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqing Xie
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dianbin Zou
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Sun
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
de Figueiredo AF, Land MGP, Ferreira GM, Mencalha A, Binato R, Capela de Matos RR, Liehr T, Silva MLM, Abdelhay E. Clinical and biological correlates of the expression of select Polycomb complex genes in Brazilian children with acute promyelocytic leukaemia. Br J Haematol 2020; 189:e245-e248. [PMID: 32314797 DOI: 10.1111/bjh.16675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/25/2020] [Accepted: 03/29/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Amanda F de Figueiredo
- Cytogenetics Laboratory, Bone Marrow Unit, National Cancer Institute (INCA), Rio de Janeiro, Brazil.,Genetics Laboratory, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marcelo G P Land
- Clinical Medicine Post-Graduation Program, College of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Gerson M Ferreira
- Stem Cell Laboratory, Bone Marrow Unit (INCA), Rio de Janeiro, Brazil
| | - Andre Mencalha
- Department of Biophysics and Biometry, Roberto Alcântara Gomes Biology Institute, Rio de Janeiro State University (UERJ), Rio de Janeiro, Brazil
| | - Renata Binato
- Stem Cell Laboratory, Bone Marrow Unit (INCA), Rio de Janeiro, Brazil
| | | | - Thomas Liehr
- Jena University Hospital, Institute of Human Genetics, Jena, Germany
| | - Maria Luiza M Silva
- Cytogenetics Laboratory, Bone Marrow Unit, National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Eliana Abdelhay
- Stem Cell Laboratory, Bone Marrow Unit (INCA), Rio de Janeiro, Brazil
| |
Collapse
|
40
|
Abdel-Khalek S, Saleh LM, Abdel-Aziz S, Hyder A, Abdel-Ghaffar H. Utility of BMI-1 and NANOG expression levels in survival prediction of pediatric acute lymphoblastic leukemia. Hematol Transfus Cell Ther 2020; 43:141-146. [PMID: 32278650 PMCID: PMC8211619 DOI: 10.1016/j.htct.2020.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/03/2020] [Accepted: 01/06/2020] [Indexed: 12/31/2022] Open
Abstract
Background Acute lymphoblastic leukemia (ALL) is the most common malignancy in children characterized by the overproduction and accumulation of immature lymphoid cells in the bone marrow and peripheral blood. The BMI-1 is an important component of the Polycomb Repressive Complex-1 (PRC1). It is an important molecule for the self-renewal of hematopoietic stem cells (HSCs). The BMI-1 expression is generally high in HSCs and decreases after cell differentiation. The BMI-1 is required for the maintenance of normal and cancer stem cells and has been reported as an oncogene in various tumors. The NANOG is a homeodomain transcription factor responsible for maintaining the stem cell compartment at the blastocyst stage of developing embryos. The NANOG gene has been proven to be transcribed in CD34+ cells and different leukemic cells. Methods The ribonucleic acid (RNA) was extracted from the peripheral blood mononuclear cells (PBMNCs) of 30 pediatric ALL patients (16 B-ALL and 14 T-ALL) and 14 healthy controls. The Bmi-1 and NANOG expression levels were determined using the quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). Results Compared to normal controls, patients with ALL exhibited upregulated levels of Bmi-1 (p = 0.03). Patients who overexpressed Bmi-1 and NANOG displayed a significantly worse survival than low-expressing patients (hazard ratio (HR) 5.74, 95% confidence interval (CI):1.48–22, p = 0.012 and HR 3.8, 95% CI:1.009–14.3, p = 0.048, respectively). Conclusions Taken together, these data suggest that the Bmi-1 and NANOG might serve as a novel survival predictor in ALL patients. Our observation also suggests that the Bmi-1 and NANOG could serve as new therapeutic targets for treatment of pediatric ALL.
Collapse
Affiliation(s)
| | - Layla M Saleh
- Oncology Center, Mansoura University, Mansoura, Egypt; Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Sherin Abdel-Aziz
- Oncology Center, Mansoura University, Mansoura, Egypt; Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ayman Hyder
- Faculty of Science, Damietta University, Damietta, Egypt
| | - Hasan Abdel-Ghaffar
- Oncology Center, Mansoura University, Mansoura, Egypt; Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
41
|
Yegin ZA, Can F, Aydın Kaynar L, Gökçen S, Eren Sadioğlu R, Özkurt ZN, Karacaoğlu Ö. Pre-transplant sTIM-3 levels may have a predictive impact on transplant outcome in acute leukemia patients. ACTA ACUST UNITED AC 2020; 25:125-133. [PMID: 32153257 DOI: 10.1080/16078454.2020.1738097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Objectives: T-cell immunoglobulin and mucin domain-containing protein-3 (TIM-3) is considered as a negative regulator of T-cell driven immune response. This study is planned to investigate the prognostic role of pre-transplant soluble TIM-3 (sTIM-3) levels in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. Methods: Pre-transplant serum sTIM-3 levels were measured in 177 allo-HSCT recipients [median age: 36(16-66) years; male/female: 111/66]. Results: Pre-transplant sTIM-3 levels were significantly higher in acute myeloid leukemia (AML) patients compared to acute lymphoblastic leukemia (ALL) patients (p = 0.01). Pre-transplant sTIM-3 levels were significantly lower in patients with abnormal cytogenetics (p = 0.017). Pre-transplant sTIM-3 levels were significantly higher in patients who developed viral hemorrhagic cystitis (p = 0.034). A positive correlation was demonstrated between sTIM-3 levels and acute graft versus host disease (GvHD) grade (p = 0.013; r = 0.299). Overall survival (OS) was not statistically different between low- and high-TIM-3 groups (%35.2 vs %20.4; p > 0.05). Primary diagnosis (p = 0.042), sinusoidal obstruction syndrome (p < 0.001), acute GvHD (p = 0.001), chronic GvHD (p = 0.009) and post-transplant relapse (p = 0.003) represented significant impact on OS. Discussion: Increased sTIM-3 levels in AML patients seem to be compatible with the previous reports. The inhibitor role of TIM-3 in cellular immune response may be a possible explanation for the association of sTIM-3 with viral infections and GvHD. However, the main challenge remains to be the ambiguous association of pre-transplant sTIM-3 levels and post-transplant complications, as allo-HSCT recipients are expected to represent donor genetic features in the post-transplant setting. Conclusion: Further studies are warranted to clarify the particular role of sTIM-3 in the allo-HSCT setting.
Collapse
Affiliation(s)
- Zeynep Arzu Yegin
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Ferda Can
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Lale Aydın Kaynar
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Sanem Gökçen
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Rezzan Eren Sadioğlu
- Department of Internal Medicine, Gazi University School of Medicine, Ankara, Turkey
| | - Zübeyde Nur Özkurt
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| | - Özlem Karacaoğlu
- Department of Hematology, Gazi University School of Medicine, Ankara, Turkey
| |
Collapse
|
42
|
Sami SA, Darwish NHE, Barile ANM, Mousa SA. Current and Future Molecular Targets for Acute Myeloid Leukemia Therapy. Curr Treat Options Oncol 2020; 21:3. [PMID: 31933183 DOI: 10.1007/s11864-019-0694-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OPINION STATEMENT Acute myeloid leukemia (AML) disease prognosis is poor and there is a high risk of chemo-resistant relapse for both young and old patients. Thus, there is a demand for alternative and target-specific drugs to improve the 5-year survival rate. Current treatment mainstays include chemotherapy, or mutation-specific targeting molecules including FLT3 inhibitors, IDH inhibitors, and monoclonal antibodies. Efforts to devise new, targeted therapy have included recent advances in methods for high-throughput genomic screening and the availability of computer-assisted techniques for the design of novel agents predicted to specifically inhibit mutant molecules involved in leukemogenesis. Crosstalk between the leukemia cells and the bone marrow microenvironment through cell surface molecules, such as the integrins αvβ3 and αvβ5, might influence drug response and AML progression. This review article focuses on current AML treatment options, new AML targeted therapies, the role of integrins in AML progression, and a potential therapeutic agent-integrin αvβ3 antagonist.
Collapse
Affiliation(s)
- Shaheedul A Sami
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA
| | - Noureldien H E Darwish
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA.,Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amanda N M Barile
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY, USA.
| |
Collapse
|
43
|
A Novel Tissue and Stem Cell Specific TERF1 Splice Variant Is Downregulated in Tumour Cells. Int J Mol Sci 2019; 21:ijms21010085. [PMID: 31877678 PMCID: PMC6981981 DOI: 10.3390/ijms21010085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/17/2022] Open
Abstract
In this study, we describe the identification of a novel splice variant of TERF1/PIN2, one of the main components of the telomeric shelterin complex. This new splice variant is identical to TERF1, apart from a 30 amino acid internal insertion near to the C-terminus of TERF1. Based on genome comparison analyses and RNA expression data, we show that this splice variant is conserved among hominidae but absent from all other species. RNA expression and histological analyses show specific expression in human spermatogonial and hematopoietic stem cells (HSCs), while all other analyzed tissues lack the expression of this TERF1-isoform, hence the name TERF1-tsi (TERF1-tissue-specific-isoform). In addition, we could not detect any expression in primary human cells and established cancer cell lines. Immunohistochemistry results involving two new rabbit polyclonal antibodies, generated against TERF1-tsi specific peptides, indicate nuclear localization of TERF1-tsi in a subset of spermatogonial stem cells. In line with this observation, immunofluorescence analyzes in various cell lines consistently revealed that ectopic TERF1-tsi localizes to the cell nucleus, mainly but not exclusively at telomeres. In a first attempt to evaluate the impact of TERF1-tsi in the testis, we have tested its expression in normal testis samples versus matched tumor samples from the same patients. Both RT-PCR and IHC show a specific downregulation of TERF1-tsi in tumor samples while the expression of TERF1 and PIN2 remains unchanged.
Collapse
|
44
|
Dao FT, Yang L, Wang YZ, Chang Y, Jiang Q, Jiang H, Liu YR, Huang XJ, Qin YZ. [Characteristic and prognostic significance of leukemia stem cells associated antigens expressions in t (8;21) acute myeloid leukemia]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:831-836. [PMID: 31775482 PMCID: PMC7364990 DOI: 10.3760/cma.j.issn.0253-2727.2019.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Indexed: 11/05/2022]
Abstract
Objective: To investigate the characteristic and prognostic significance of leukemia stem cells associated antigens expressions including CD34, CD38, CD123, CD96 and TIM-3 in t (8;21) AML. Methods: Bone marrow samples of 47 t (8;21) AML patients were collected at diagnosis from October 2015 to April 2018 in Peking University Peoples' Hospital, then flow cytometry method was performed to detect the expression frequencies of CD34, CD38, CD123, CD96 and TIM-3 to analyze the relationship between leukemia stem cells associated antigens expressions and relapse. Results: Of 47 t (8;21) AML patients tested, the median percentages of CD34(+)CD38(-), CD34(+) CD38(-)CD123(+), CD34(+)CD38(-) CD96(+) and CD34(+) CD38(-) TIM-3(+) cells among nucleated cells were 2.37%, 0.24%, 0.27% and 0.06%, respectively. All the frequencies of CD34(+)CD38(-), CD34(+)CD38(-)CD123(+), CD34(+)CD38(-)CD96(+) and CD34(+) CD38(-)TIM-3(+) cells had no impact on the achievement of CR after the first course of induction. All higher frequencies of CD34(+)CD38(-), CD34(+)CD38(-)CD123(+), CD34(+)CD38(-)CD96(+) cells were related to higher 2-year CIR rate. Whereas, the frequency of CD34(+) CD38(-) TIM-3(+) cells had no impact on CIR rate. Both high frequency of CD34(+) CD38(-) cells and the high level of minimal residual diseases (patients with <3-log reduction in the RUNX1-RUNX1T1 transcript level after the second consolidation therapy) were independent poor prognostic factors of CIR[P=0.025, HR=6.9 (95%CI 1.3-37.4) ; P=0.031, HR=11.1 (95%CI 1.2-99.2) ]. Conclusion: Different leukemia stem cells associated antigens had distinct prognostic significance in t (8;21) AML. High frequencies of CD34(+) CD38(-), CD34(+) CD38(-) CD123(+) and CD34(+)CD38(-)CD96(+) cells at diagnosis predicted relapse in patients with t (8;21) AML.
Collapse
Affiliation(s)
- F T Dao
- Peking University Peoples'Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Darwish NHE, Sudha T, Godugu K, Bharali DJ, Elbaz O, El-Ghaffar HAA, Azmy E, Anber N, Mousa SA. Novel Targeted Nano-Parthenolide Molecule against NF-kB in Acute Myeloid Leukemia. Molecules 2019; 24:molecules24112103. [PMID: 31163672 PMCID: PMC6600366 DOI: 10.3390/molecules24112103] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/30/2022] Open
Abstract
The targeted nano-encapsulation of anticancer drugs can improve drug delivery and the selective targeting of cancer cells. Nuclear factor kappa B (NF-kB) is a regulator for different biological responses, including cell proliferation and differentiation. In acute myeloid leukemia (AML), constitutive NF-κB has been detected in more than 50% of cases, enabling leukemic cells to resist apoptosis and stimulate uncontrolled proliferation. We evaluated NF-kB expression in bone marrow samples from 103 patients with AML using quantitative real time polymerase chain reaction (RT-PCR) and found that expression was increased in 80.5% (83 out 103) of these patients with AML in comparison to the control group. Furthermore, overexpressed transmembrane glycoprotein (CD44) on leukemic cells in comparison to normal cells is known to play an important role in leukemic cell engraftment and survival. We designed poly lactide co-glycolide (PLGA) nanoparticles conjugated with antiCD44 and encapsulating parthenolide (PTL), a nuclear factor kappa B (NF-kB) inhibitor, in order to improve the selectivity and targeting of leukemic cells and to spare normal cells. In vitro, in leukemic cell lines Kasumi-1, KG-1a, and THP-1, proliferation was decreased by 40% (** p < 0.01) with 5 µM PLGA-antiCD44-PTL nanoparticles in comparison to the same concentration of free PTL (~10%). The higher uptake of the nanoparticles by leukemic cells was confirmed with confocal microscopy. In conclusion, PLGA-antiCD44-PTL nanoparticles improved the bioavailability and selective targeting of leukemic cells, thus holding promise as a drug delivery system to improve the cure rate of AML.
Collapse
Affiliation(s)
- Noureldien H E Darwish
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Thangirala Sudha
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Kavitha Godugu
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Dhruba J Bharali
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Osama Elbaz
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Hasan A Abd El-Ghaffar
- Hematology Unit, Clinical Pathology Department, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Emad Azmy
- Clinical Hematology Unit, Mansoura University Oncology Center, Mansoura University, Mansoura 35516, Egypt.
| | - Nahla Anber
- Fellow of Biochemistry Emergency Hospital, Mansoura University, Mansoura 35516, Egypt.
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
46
|
Acute Myeloid Leukemia Stem Cell Heterogeneity and Its Clinical Relevance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1139:153-169. [DOI: 10.1007/978-3-030-14366-4_9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
47
|
Ma H, Padmanabhan IS, Parmar S, Gong Y. Targeting CLL-1 for acute myeloid leukemia therapy. J Hematol Oncol 2019; 12:41. [PMID: 31014360 PMCID: PMC6480870 DOI: 10.1186/s13045-019-0726-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 03/27/2019] [Indexed: 02/05/2023] Open
Abstract
Despite major scientific discoveries and novel therapies over the past four decades, the treatment outcomes of acute myeloid leukemia (AML), especially in the adult patient population remain dismal. In the past few years, an increasing number of targets such as CD33, CD123, CLL-1, CD47, CD70, and TIM3, have been developed for immunotherapy of AML. Among them, CLL-1 has attracted the researchers’ attention due to its high expression in AML while being absent in normal hematopoietic stem cell. Accumulating evidence have demonstrated CLL-1 is an ideal target for AML. In this paper, we will review the expression of CLL-1 on normal cells and AML, the value of CLL-1 in diagnosis and follow-up, and targeting CLL-1 therapy-based antibody and chimeric antigen receptor T cell therapy as well as providing an overview of CLL-1 as a target for AML.
Collapse
Affiliation(s)
- Hongbing Ma
- Hematology Department, West China Hospital, Sichuan University, Chengdu, China
| | | | - Simrit Parmar
- Department of Lymphoma and Myeloma, MD Anderson Cancer Center, Texas University, Houston, USA.
| | - Yuping Gong
- Hematology Department, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
48
|
Paubelle E, Rocher C, Julia E, Thomas X. Chimeric Antigen Receptor-Engineered T Cell Therapy in Acute Myeloid Leukaemia. EUROPEAN MEDICAL JOURNAL 2018. [DOI: 10.33590/emj/10314141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Acute myeloid leukaemia (AML) is a disease with a very poor outcome and remains an area of significant unmet need, necessitating novel therapeutic strategies. The progress made in the field of immunotherapy, in particular chimeric antigen receptor (CAR)-engineered T cells, has given rise to many hopes for pathologies such as B cell acute lymphoblastic leukaemia and B cell lymphoma, and many studies have attempted to translate these successes to AML. This review summarises the recent advances in, and defines an ideal target for, CAR T cell therapy in AML.
Collapse
Affiliation(s)
- Etienne Paubelle
- Department of Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France; LBMC, ENS, CNRS UMR5239, Faculté de Médecine Lyon-Sud, Lyon, France
| | - Clément Rocher
- Department of Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
| | - Edith Julia
- Department of Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
| | - Xavier Thomas
- Department of Hematology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
| |
Collapse
|
49
|
Zheng B, Yu SF, Del Rosario G, Leong SR, Lee GY, Vij R, Chiu C, Liang WC, Wu Y, Chalouni C, Sadowsky J, Clark V, Hendricks A, Poon KA, Chu W, Pillow T, Schutten MM, Flygare J, Polson AG. An Anti-CLL-1 Antibody-Drug Conjugate for the Treatment of Acute Myeloid Leukemia. Clin Cancer Res 2018; 25:1358-1368. [PMID: 29959143 DOI: 10.1158/1078-0432.ccr-18-0333] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/07/2018] [Accepted: 06/25/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE The treatment of acute myeloid leukemia (AML) has not significantly changed in 40 years. Cytarabine- and anthracycline-based chemotherapy induction regimens (7 + 3) remain the standard of care, and most patients have poor long-term survival. The reapproval of Mylotarg, an anti-CD33-calicheamicin antibody-drug conjugate (ADC), has demonstrated ADCs as a clinically validated option to enhance the effectiveness of induction therapy. We are interested in developing a next-generation ADC for AML to improve upon the initial success of Mylotarg. EXPERIMENTAL DESIGN The expression pattern of CLL-1 and its hematopoietic potential were investigated. A novel anti-CLL-1-ADC, with a highly potent pyrrolobenzodiazepine (PBD) dimer conjugated through a self-immolative disulfide linker, was developed. The efficacy and safety profiles of this ADC were evaluated in mouse xenograft models and in cynomolgus monkeys. RESULTS We demonstrate that CLL-1 shares similar prevalence and trafficking properties that make CD33 an excellent ADC target for AML, but lacks expression on hematopoietic stem cells that hampers current CD33-targeted ADCs. Our anti-CLL-1-ADC is highly effective at depleting tumor cells in AML xenograft models and lacks target independent toxicities at doses that depleted target monocytes and neutrophils in cynomolgus monkeys. CONCLUSIONS Collectively, our data suggest that an anti-CLL-1-ADC has the potential to become an effective and safer treatment for AML in humans, by reducing and allowing for faster recovery from initial cytopenias than the current generation of ADCs for AML.
Collapse
Affiliation(s)
- Bing Zheng
- Research and Early Development, Genentech Inc., South San Francisco, California.
| | - Shang-Fan Yu
- Research and Early Development, Genentech Inc., South San Francisco, California
| | | | - Steven R Leong
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Genee Y Lee
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Rajesh Vij
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Cecilia Chiu
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Wei-Ching Liang
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Yan Wu
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Cecile Chalouni
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Jack Sadowsky
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Vanessa Clark
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Angela Hendricks
- Research and Early Development, Genentech Inc., South San Francisco, California
| | | | - Wayne Chu
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Thomas Pillow
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Melissa M Schutten
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - John Flygare
- Research and Early Development, Genentech Inc., South San Francisco, California
| | - Andrew G Polson
- Research and Early Development, Genentech Inc., South San Francisco, California.
| |
Collapse
|
50
|
Maeda A, Nishida Y, Weetall M, Cao L, Branstrom A, Ishizawa J, Nii T, Schober WD, Abe Y, Matsue K, Yoshimura M, Kimura S, Kojima K. Targeting of BMI-1 expression by the novel small molecule PTC596 in mantle cell lymphoma. Oncotarget 2018; 9:28547-28560. [PMID: 29983879 PMCID: PMC6033370 DOI: 10.18632/oncotarget.25558] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/21/2018] [Indexed: 12/24/2022] Open
Abstract
Despite the development of the novel Bruton tyrosine kinase inhibitor ibrutinib, mantle cell lymphoma (MCL) remains an incurable B-cell non-Hodgkin lymphoma. BMI-1 is required for the self-renewal and maintenance of MCL-initiating stem cells. Upregulation of BMI-1 has been reported in MCL patients, especially in those with refractory/relapsed disease. We studied the effects of a novel small-molecule selective inhibitor of BMI1 expression, PTC596, in MCL cells. Eight MCL cell lines and patient-derived samples were exposed to PTC596. PTC596 induced mitochondrial apoptosis, as evidenced by loss of mitochondrial membrane potential, caspase-3 cleavage, BAX activation, and phosphatidylserine externalization. There was a positive correlation between baseline BMI-1 protein levels and PTC596-induced apoptosis. p53 status did not affect sensitivity to PTC596. PTC596 effectively decreased BMI-1-expressing and tumor-initiating side population MCL cells (IC50: 138 nM) compared with ibrutinib, which modestly decreased side population cells. Interestingly, PTC596, reported to target cancer stem cells, decreased MCL-1 expression levels and antagonized ibrutinib-induced increase in MCL-1 expression, leading to synergistic apoptosis induction in MCL cells. There are currently no drugs that specifically target cancer stem cell fractions, and a reduction in BMI-1 protein by PTC596 may offer a novel therapeutic strategy for MCL.
Collapse
Affiliation(s)
- Aya Maeda
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Yuki Nishida
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | | | | | | | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Takenobu Nii
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy D Schober
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yoshiaki Abe
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Mariko Yoshimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| | - Kensuke Kojima
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Saga University, Saga, Japan
| |
Collapse
|