1
|
Pan S, Cai Q, Wei Y, Tang H, Zhang Y, Zhou W, Deng T, Mo W, Wang S, Wang C, Chen C. Increased co-expression of ICOS and PD-1 predicts poor overall survival in patients with acute myeloid leukemia. Immunobiology 2024; 229:152804. [PMID: 38615511 DOI: 10.1016/j.imbio.2024.152804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Inducible co-stimulatory factor (ICOS) has a dual role: activating cytotoxic T cells against tumors or exacerbating immunosuppression of regulatory T cells (Tregs) to participate in immune evasion. However, the correlation between ICOS and its co-expression with inhibitory immune checkpoints (IICs) and prognosis in acute myeloid leukemia (AML) is little known. METHODS The prognostic importance of ICOS and IICs in 62 bone marrow (BM) samples of de novo AML patients from our clinical center (GZFPH) was explored and then the RNA sequencing data of 155 AML patients from the Cancer Genome Atlas (TCGA) database was used for validation. RESULTS In both GZFPH and TCGA cohorts, high expression of ICOS was significantly associated with poor overall survival (OS) in patients with AML (P < 0.05). Importantly, co-expression of ICOS and PD-1, PD-L1, PD-L2, CTLA-4, and LAG-3 predicted poor OS in AML; among them, ICOS/PD-1 was the optimal combination of immune checkpoints (ICs). The co-expression of ICOS and PD-1 was correlated with poor OS in non-acute promyelocytic leukemia (non-APL) patients following chemotherapy. Additionally, ICOS/PD-1 was an independent OS-predicting factor (P < 0.05). Notably, a nomogram model was constructed by combining ICOS/PD-1, age, European Leukemia Net (ELN) risk stratification, and therapy to visually and personalized predict the 1-, 3-, and 5-year OS of patients with non-APL. CONCLUSION Increased expression of ICOS predicted poor outcomes, and ICOS/PD-1 was the optimal combination of ICs to predict outcomes in patients with AML, which might be a potential immune biomarker for designing novel AML therapy.
Collapse
Affiliation(s)
- Shiyi Pan
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Qinghua Cai
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Yiqiong Wei
- Department of Neurology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Haifeng Tang
- Department of Surgery, The Third School of Clinical Medicine, Southern Medical University, Guangzhou 516006, China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Wei Zhou
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Tingfen Deng
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| | - Caixia Wang
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| | - Cunte Chen
- Department of Hematology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510180, China.
| |
Collapse
|
2
|
Hazrati A, Malekpour K, Khorramdelazad H, Rajaei S, Hashemi SM. Therapeutic and immunomodulatory potentials of mesenchymal stromal/stem cells and immune checkpoints related molecules. Biomark Res 2024; 12:35. [PMID: 38515166 PMCID: PMC10958918 DOI: 10.1186/s40364-024-00580-2] [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: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are used in many studies due to their therapeutic potential, including their differentiative ability and immunomodulatory properties. These cells perform their therapeutic functions by using various mechanisms, such as the production of anti-inflammatory cytokines, growth factors, direct cell-to-cell contact, extracellular vesicles (EVs) production, and mitochondrial transfer. However, mechanisms related to immune checkpoints (ICPs) and their effect on the immunomodulatory ability of MSCs are less discussed. The main function of ICPs is to prevent the initiation of unwanted responses and to regulate the immune system responses to maintain the homeostasis of these responses. ICPs are produced by various types of immune system regulatory cells, and defects in their expression and function may be associated with excessive responses that can ultimately lead to autoimmunity. Also, by expressing different types of ICPs and their ligands (ICPLs), tumor cells prevent the formation and durability of immune responses, which leads to tumors' immune escape. ICPs and ICPLs can be produced by MSCs and affect immune cell responses both through their secretion into the microenvironment or direct cell-to-cell interaction. Pre-treatment of MSCs in inflammatory conditions leads to an increase in their therapeutic potential. In addition to the effect that inflammatory environments have on the production of anti-inflammatory cytokines by MSCs, they can increase the expression of various types of ICPLs. In this review, we discuss different types of ICPLs and ICPs expressed by MSCs and their effect on their immunomodulatory and therapeutic potential.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Samira Rajaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Unterfrauner M, Rejeski HA, Hartz A, Bohlscheid S, Baudrexler T, Feng X, Rackl E, Li L, Rank A, Filippini Velázquez G, Schmid C, Schmohl J, Bojko P, Schmetzer H. Granulocyte-Macrophage-Colony-Stimulating-Factor Combined with Prostaglandin E1 Create Dendritic Cells of Leukemic Origin from AML Patients' Whole Blood and Whole Bone Marrow That Mediate Antileukemic Processes after Mixed Lymphocyte Culture. Int J Mol Sci 2023; 24:17436. [PMID: 38139264 PMCID: PMC10743754 DOI: 10.3390/ijms242417436] [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: 10/31/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Although several (chemotherapeutic) protocols to treat acute myeloid leukemia (AML) are available, high rates of relapses in successfully treated patients occur. Strategies to stabilize remissions are greatly needed. The combination of the (clinically approved) immune-modulatory compounds Granulocyte-Macrophage-Colony-Stimulating-Factor (GM-CSF) and Prostaglandine E1 (PGE-1) (Kit-M) converts myeloid blasts into dendritic cells of leukemic origin (DCleu). After stimulation with DCleu ex vivo, leukemia-specific antileukemic immune cells are activated. Therefore, Kit-M treatment may be an attractive immunotherapeutic tool to treat patients with myeloid leukemia. Kit-M-mediated antileukemic effects on whole bone marrow (WBM) were evaluated and compared to whole blood (WB) to evaluate the potential effects of Kit-M on both compartments. WB and WBM samples from 17 AML patients at first diagnosis, in persisting disease and at relapse after allogeneic stem cell transplantation (SCT) were treated in parallel with Kit-M to generate DC/DCleu. Untreated samples served as controls. After a mixed lymphocyte culture enriched with patients' T cells (MLC), the leukemia-specific antileukemic effects were assessed through the degranulation- (CD107a+ T cells), the intracellular IFNγ production- and the cytotoxicity fluorolysis assay. Quantification of cell subtypes was performed via flow cytometry. In both WB and WBM significantly higher frequencies of (mature) DCleu were generated without induction of blast proliferation in Kit-M-treated samples compared to control. After MLC with Kit-M-treated vs. not pretreated WB or WBM, frequencies of (leukemia-specific) immunoreactive cells (e.g., non-naive, effector-, memory-, CD3+β7+ T cells, NK- cells) were (significantly) increased, whereas leukemia-specific regulatory T cells (Treg, CD152+ T cells) were (significantly) decreased. The cytotoxicity fluorolysis assay showed a significantly improved blast lysis in Kit-M-treated WB and WBM compared to control. A parallel comparison of WB and WBM samples revealed no significant differences in frequencies of DCleu, (leukemia-specific) immunoreactive cells and achieved antileukemic processes. Kit-M was shown to have comparable effects on WB and WBM samples regarding the generation of DCleu and activation of (antileukemic) immune cells after MLC. This was true for samples before or after SCT. In summary, a potential Kit-M in vivo treatment could lead to antileukemic effects in WB as well as WBM in vivo and to stabilization of the disease or remission in patients before or after SCT. A clinical trial is currently being planned.
Collapse
Affiliation(s)
| | - Hazal Aslan Rejeski
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Anne Hartz
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Sophia Bohlscheid
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Tobias Baudrexler
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Xiaojia Feng
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Elias Rackl
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Lin Li
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| | - Andreas Rank
- Department of Hematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | | | - Christoph Schmid
- Department of Hematology and Oncology, University Hospital of Augsburg, 86156 Augsburg, Germany
| | - Jörg Schmohl
- Department of Hematology and Oncology, Diakonieklinikum Stuttgart, 70176 Stuttgart, Germany
| | - Peter Bojko
- Department of Hematology and Oncology, Rotkreuzklinikum Munich, 80634 Munich, Germany
| | - Helga Schmetzer
- Department of Medicine III, University Hospital of Munich, 81377 Munich, Germany
| |
Collapse
|
4
|
Wang P, Zhang Y, Cai Q, Long Q, Pan S, Zhou W, Deng T, Mo W, Wang S, Zhang Y, Wang C, Chen C. Optimal combination of immune checkpoint and senescence molecule predicts adverse outcomes in patients with acute myeloid leukemia. Ann Med 2023; 55:2201507. [PMID: 37070487 PMCID: PMC10120552 DOI: 10.1080/07853890.2023.2201507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND High expression of immune checkpoints (ICs) and senescence molecules (SMs) contributes to T cell dysfunction, tumor escape, and progression, but systematic evaluation of them in co-expression patterns and prognosis in acute myeloid leukemia (AML) was lacking. METHODS Three publicly available datasets (TCGA, Beat-AML, and GSE71014) were first used to explore the effect of IC and SM combinations on prognosis and the immune microenvironment in AML, and bone marrow samples from 68 AML patients from our clinical center (GZFPH) was further used to validate the findings. RESULTS High expression of CD276, Bcl2-associated athanogene 3 (BAG3), and SRC was associated with poor overall survival (OS) of AML patients. CD276/BAG3/SRC combination, standard European Leukemia Net (ELN) risk stratification, age, and French-American-British (FAB) subtype were used to construct a nomogram model. Interestingly, the new risk stratification derived from the nomogram was better than the standard ELN risk stratification in predicting the prognosis for AML. A weighted combination of CD276 and BAG3/SRC positively corrected with TP53 mutation, p53 pathway, CD8+ T cells, activated memory CD4+ T cells, T-cell senescence score, and Tumor Immune Dysfunction and Exclusion (TIDE) score estimated by T-cell dysfunction. CONCLUSION High expression of ICs and SMs was associated with poor OS of AML patients. The co-expression patterns of CD276 and BAG3/SRC might be potential biomarkers for risk stratification and designing combinational immuno-targeted therapy in AML.Key MessagesHigh expression of CD276, BAG3, and SRC was associated with poor overall survival of AML patients.The co-expression patterns of CD276 and BAG3/SRC might be potential biomarkers for risk stratification and designing combinational immuno-targeted therapy in AML.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Oncology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R. China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, P.R. China
| | - Yuling Zhang
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Qinghua Cai
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Qingqin Long
- Department of Oncology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R. China
| | - Shiyi Pan
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Wei Zhou
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Tingfen Deng
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Wenjian Mo
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
- Yuping Zhang Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou510180, P.R. China
| | - Caixia Wang
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
- Caixia Wang
| | - Cunte Chen
- Department of Hematology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, P.R.China
- CONTACT Cunte Chen
| |
Collapse
|
5
|
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
|
6
|
Chen M, Fu M, Gong M, Gao Y, Wang A, Zhao W, Wu X, Wang H. Twenty-four-color full spectrum flow cytometry panel for minimal residual disease detection in acute myeloid leukemia. Open Med (Wars) 2023; 18:20230745. [PMID: 37533738 PMCID: PMC10390751 DOI: 10.1515/med-2023-0745] [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: 12/29/2022] [Revised: 05/22/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023] Open
Abstract
Full spectrum flow cytometry brings a breakthrough for minimal residual disease (MRD) detection in acute myeloid leukemia (AML). We aimed to explore the role of a new panel in MRD detection. We established a 24-color full-spectrum flow cytometry panel. A tube of 24-color antibodies included CD45, CD117, CD34, HLA-DR, CD15, CD64, CD14, CD11c, CD11b, CD13, CD33, CD371, CD7, CD56, CD19, CD4, CD2, CD123, CD200, CD38, CD96, CD71, CD36, and CD9. We discovered that when a tube meets 26 parameters (24 colors), these markers were not only limited to the observation of MRD in AML, but also could be used for fine clustering of bone marrow cells. Mast cells, basophils, myeloid dendritic cells, and plasmacoid dendritic cells were more clearly observed. In addition, immune checkpoint CD96 had the higher expression in CD117+ myeloid naive cells and CD56dimNK cells, while had the lower expression in CD56briNK cells in AML-MRD samples than in normal bone marrow samples. CD200 expression was remarkably enhanced in CD117+ myeloid naive cells, CD4+ T cells, T cells, activated T cells, CD56dimNK cells, and CD56briNK cells in AML-MRD samples. Our results can be used as important basis for auxiliary diagnosis, prognosis judgment, treatment guidance, and immune regulation in AML.
Collapse
Affiliation(s)
- Man Chen
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Sanhe, Langfang, Hebei, China
| | - Minjing Fu
- Department of Laboratory Medicine, Beijing Ludaopei Hospital, Beijing, China
| | - Meiwei Gong
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Sanhe, Langfang, Hebei, China
| | - Yajing Gao
- Cytek (Shanghai) BioSciences Co. Ltd, Shanghai, China
| | - Aixian Wang
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Sanhe, Langfang, Hebei, China
| | - Wei Zhao
- Department of Stem Cell Transplantation, Beijing Ludaopei Hospital, Beijing, China
| | - Xueying Wu
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Sanhe, Langfang, Hebei, China
| | - Hui Wang
- Department of Laboratory Medicine, Hebei Yanda Ludaopei Hospital, Sanhe, Langfang, Hebei, China
| |
Collapse
|
7
|
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
|
8
|
Ji R, Li Y, Huang R, Xiong J, Wang X, Zhang X. Recent Advances and Research Progress in Biomarkers for Chronic Graft Versus Host Disease. Crit Rev Oncol Hematol 2023; 186:103993. [PMID: 37061073 DOI: 10.1016/j.critrevonc.2023.103993] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023] Open
Abstract
Chronic graft-versus host disease (cGVHD) is a major risk for patients undergoing allogeneic hematopoietic stem cell transplantation. With the emergence of novel therapies and the increased understanding of the mechanisms underlying cGVHD, there are more options for cGVHD treatment. Regardless of improvements in treatment, diagnosis mainly depends on identification of symptoms, which makes precise treatment a challenge. Numerous biomarkers for cGVHD have been validated and have demonstrated strong associations with prognosis and response to treatment. The most common biomarkers mainly include critical types of immune cells, chemokines, cytokines, microRNAs, and autoantibodies, all of which play important roles in the development of cGVHD. Compared to traditional tools, biomarkers have several advantages, for example, they can be applied for early diagnosis, to identify cGVHD risk before onset, and predict which therapy is most likely to benefit patients. In this review, we summarize biomarkers with potential clinical value and discuss future applications.
Collapse
Affiliation(s)
- Rui Ji
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Yue Li
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Jingkang Xiong
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing 400037, China; Jinfeng Laboratory, Chongqing 400037, China.
| |
Collapse
|
9
|
Zhang Q, Ma R, Chen H, Guo W, Li Z, Xu K, Chen W. CD86 Is Associated with Immune Infiltration and Immunotherapy Signatures in AML and Promotes Its Progression. JOURNAL OF ONCOLOGY 2023; 2023:9988405. [PMID: 37064861 PMCID: PMC10104747 DOI: 10.1155/2023/9988405] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/21/2022] [Accepted: 07/25/2022] [Indexed: 04/18/2023]
Abstract
Background Cluster of differentiation 86 (CD86), also known as B7-2, is a molecule expressed on antigen-presenting cells that provides the costimulatory signals required for T cell activation and survival. CD86 binds to two ligands on the surface of T cells: the antigen CD28 and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). By binding to CD28, CD86-together with CD80-promotes the participation of T cells in the antigen presentation process. However, the interrelationships among CD86, immunotherapy, and immune infiltration in acute myeloid leukemia (AML) are unclear. Methods The immunological effects of CD86 in various cancers (including on chemokines, immunostimulators, MHC, and receptors) were evaluated through a pan-cancer analysis using TCGA and GEO databases. The relationship between CD86 expression and mononucleotide variation, gene copy number variation, methylation, immune checkpoint blockers (ICBs), and T-cell inflammation score in AML was subsequently examined. ESTIMATE and limma packages were used to identify genes at the intersection of CD86 with StromalScore and ImmuneScore. Subsequently, GO/KEGG and PPI network analyses were performed. The immune risk score (IRS) model was constructed, and the validation set was used for verification. The predictive value was compared with the TIDE score. Results CD86 was overexpressed in many cancers, and its overexpression was associated with a poor prognosis. CD86 expression was positively correlated with the expression of CTLA4, PDCD1LG2, IDO1, HAVCR2, and other genes and negatively correlated with CD86 methylation. The expression of CD86 in AML cell lines was detected by QRT-PCR and Western blot, and the results showed that CD86 was overexpressed in AML cell lines. Immune infiltration assays showed that CD86 expression was positively correlated with CD8 T cell, Dendritic cell, macrophage, NK cell, and Th1_cell and also with immune examination site, immune regulation, immunotherapy response, and TIICs. ssGSEA showed that CD86 was enriched in immune-related pathways, and CD86 expression was correlated with mutations in the genes RB1, ERBB2, and FANCC, which are associated with responses to radiotherapy and chemotherapy. The IRS score performed better than the TIDE website score. Conclusion CD86 appears to participate in immune invasion in AML and is an important player in the tumor microenvironment in this malignancy. At the same time, the IRS score developed by us has a good effect and may provide some support for the diagnosis of AML. Thus, CD86 may serve as a potential target for AML immunotherapy.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ruixue Ma
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huimin Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wentong Guo
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhenyu Li
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
| | - Wei Chen
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, Jiangsu, China
- Department of Hematology, The First People's Hospital of Suqian, Suqian, Jiangsu, China
| |
Collapse
|
10
|
Present and Future Role of Immune Targets in Acute Myeloid Leukemia. Cancers (Basel) 2022; 15:cancers15010253. [PMID: 36612249 PMCID: PMC9818182 DOI: 10.3390/cancers15010253] [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: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
It is now well known that the bone marrow (BM) cell niche contributes to leukemogenesis, but emerging data support the role of the complex crosstalk between AML cells and the BM microenvironment to induce a permissive immune setting that protects leukemic stem cells (LSCs) from therapy-induced death, thus favoring disease persistence and eventual relapse. The identification of potential immune targets on AML cells and the modulation of the BM environment could lead to enhanced anti-leukemic effects of drugs, immune system reactivation, and the restoration of AML surveillance. Potential targets and effectors of this immune-based therapy could be monoclonal antibodies directed against LSC antigens such as CD33, CD123, and CLL-1 (either as direct targets or via several bispecific T-cell engagers), immune checkpoint inhibitors acting on different co-inhibitory axes (alone or in combination with conventional AML drugs), and novel cellular therapies such as chimeric antigen receptor (CAR) T-cells designed against AML-specific antigens. Though dozens of clinical trials, mostly in phases I and II, are ongoing worldwide, results have still been negatively affected by difficulties in the identification of the optimal targets on LSCs.
Collapse
|
11
|
Zheng Y, Weng X, Hu D, He J. Identification of a signature based on non‐apoptotic regulatory cell death to improve prognosis prediction in acute myeloid leukaemia. Br J Haematol 2022; 201:95-105. [PMID: 36484284 DOI: 10.1111/bjh.18601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Although anti-apoptotic cell death is a common feature of cancer and non-apoptotic regulatory cell death (RCD) is highly correlated with cancer progression and response to therapy, its prognostic role in patients with acute myeloid leukaemia (AML) is unknown. The RNA sequence and clinical data from AML patients were downloaded from the TCGA and GEO databases. The prognostic characteristics of non-apoptotic RCD-related genes (NRGs) were determined by Cox and LASSO regression analysis. Thirteen NRG signatures were identified as independent prognostic parameters in patients with AML that outperformed other prognostic models. Higher NRG scores were associated with shorter survival and less retention of tumour mutations. Although patients with high NRG risk have abundant signalling pathways for cell adhesion, cytokine upregulation, and cellular defence responses, patients with low NRG risk may benefit the most from immunotherapy. Specifically, patients with high NRG score may benefit from treatment with anti-EGFR and CDK2 inhibitors, including erlotinib and roscovitine. The NPM1 and FLT3 mutant cell lines undergo alterations after multiple drug treatments. Our established NRG signature and scoring highlight its vital clinical significance, emphasize the inevitability of stratifying treatment for different mutation subtypes and provide new ideas to guide personalized immunotherapy strategies for AML patients.
Collapse
Affiliation(s)
- Yu Zheng
- Key State Laboratory of Medical Genomics, National Center for Translational Medicine in Shanghai, Institute of Hematology Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiangqin Weng
- Key State Laboratory of Medical Genomics, National Center for Translational Medicine in Shanghai, Institute of Hematology Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Dong Hu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jing He
- Institute of Hematology, Union Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
12
|
Zhong M, Gao R, Zhao R, Huang Y, Chen C, Li K, Yu X, Nie D, Chen Z, Liu X, Liu Z, Chen S, Lu Y, Yu Z, Wang L, Li P, Zeng C, Li Y. BET bromodomain inhibition rescues PD-1-mediated T-cell exhaustion in acute myeloid leukemia. Cell Death Dis 2022; 13:671. [PMID: 35918330 PMCID: PMC9346138 DOI: 10.1038/s41419-022-05123-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/21/2023]
Abstract
Sustained expression of programmed cell death receptor-1 (PD-1) is correlated with the exhaustion of T cells, and blockade of the PD-1 pathway is an effective immunotherapeutic strategy for treating various cancers. However, response rates are limited, and many patients do not achieve durable responses. Thus, it is important to seek additional strategies that can improve anticancer immunity. Here, we report that the bromodomain and extraterminal domain (BET) inhibitor JQ1 inhibits PD-1 expression in Jurkat T cells, primary T cells, and T-cell exhaustion models. Furthermore, JQ1 dramatically impaired the expression of PD-1 and T-cell immunoglobulin mucin-domain-containing-3 (Tim-3) and promoted the secretion of cytokines in T cells from patients with acute myeloid leukemia (AML). In line with that, BET inhibitor-treated CD19-CAR T and CD123-CAR T cells have enhanced anti-leukemia potency and resistant to exhaustion. Mechanistically, BRD4 binds to the NFAT2 and PDCD1 (encoding PD-1) promoters, and NFAT2 binds to the PDCD1 and HAVCR2 (encoding Tim-3) promoters. JQ1-treated T cells showed downregulated NFAT2, PD-1, and Tim-3 expression. In addition, BET inhibitor suppressed programmed death-ligand 1 (PD-L1) expression and cell growth in AML cell lines and in primary AML cells. We also demonstrated that JQ1 treatment led to inhibition of leukemia progression, reduced T-cell PD-1/Tim-3 expression, and prolonged survival in MLL-AF9 AML mouse model and Nalm6 (B-cell acute lymphoblastic leukemia cell)-bearing mouse leukemia model. Taken together, BET inhibition improved anti-leukemia immunity by regulating PD-1/PD-L1 expression, and also directly suppressed AML cells, which provides novel insights on the multiple effects of BET inhibition for cancer therapy.
Collapse
Affiliation(s)
- Mengjun Zhong
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Rili Gao
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Ruocong Zhao
- grid.9227.e0000000119573309Center for Cell Regeneration and Biotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, P. R. China
| | - Youxue Huang
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Cunte Chen
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Kehan Li
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Xibao Yu
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Dingrui Nie
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Zheng Chen
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Xin Liu
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Zhuandi Liu
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Shaohua Chen
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Yuhong Lu
- grid.258164.c0000 0004 1790 3548Department of Hematology, First Affiliated Hospital, Jinan University, 510632 Guangzhou, P. R. China
| | - Zhi Yu
- grid.258164.c0000 0004 1790 3548Department of Hematology, First Affiliated Hospital, Jinan University, 510632 Guangzhou, P. R. China
| | - Liang Wang
- grid.258164.c0000 0004 1790 3548Department of Oncology, First Affiliated Hospital, Jinan University, 510632 Guangzhou, P. R. China
| | - Peng Li
- grid.9227.e0000000119573309Center for Cell Regeneration and Biotherapy, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, P. R. China
| | - Chengwu Zeng
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| | - Yangqiu Li
- grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, 510632 Guangzhou, P. R. China
| |
Collapse
|
13
|
Xu Q, Guo T. Somatic mutation-associated risk index based on lncRNA expression for predicting prognosis in acute myeloid leukemia. Hematology 2022; 27:659-671. [PMID: 35666642 DOI: 10.1080/16078454.2022.2056677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Objectives: Genomic instability has several implications for acute myeloid leukemia (AML) prognosis. This article aims to construct a somatic mutation-associated risk index (SMRI) of genomic instability for AML to predict prognosis and explore the potential determinants of AML prognosis.Methods: We obtained differentially expressed lncRNAs from genomic instability subtypes and selected six lncRNAs to construct the SMRI through multivariate Cox regression analysis. The median SMRI classified patients into high and low SMRI groups. Kaplan-Meier survival analysis was used to clarify the prognostic differences of SMRI subtypes. Receiver operating characteristic curve analysis was performed to elucidate the value of SMRI as a prognostic indicator. Gene set variation analysis, tumor mutation burden (TMB) analysis, immune infiltration, and immune checkpoint expression analysis were performed to investigate possible causes for the differences in prognosis of SMRI subtypes.Results: The high SMRI group exhibited a poor prognosis, which was characterized by elevated levels of TMB, mutation counts (TP53, NPM1, DNMT3A, and FLT3-TKD), CD8+ T cell infiltration, and immune checkpoint (PD-1, PD-L2, CTLA4, LAG3) expression. The SMRI was still associated with prognosis, even after adjustment for age, sex, cytogenetic risk, DNMT3A status, FLT3 status, and NPM1 status. Gene set variation analysis showed that AML with FLT3-ITD mutation, CEBPA mutation, and LSCs (leukemia stem cells) were enriched in the high SMRI group.Conclusion: Our research suggests that the SMRI derived from genomic instability subtypes is a useful biomarker for predicting prognosis and may be beneficial for improving the clinical outcome of patients with AML.
Collapse
Affiliation(s)
- Qiang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
14
|
Jiang Z, Long J, Deng K, Zheng Y, Chen M. eRNAs Identify Immune Microenvironment Patterns and Provide a Novel Prognostic Tool in Acute Myeloid Leukemia. Front Mol Biosci 2022; 9:877117. [PMID: 35586193 PMCID: PMC9108177 DOI: 10.3389/fmolb.2022.877117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/28/2022] [Indexed: 12/19/2022] Open
Abstract
Background: Enhancer RNAs (eRNAs) play an essential role in tumorigenesis as non-coding RNAs transcribed from enhancer regions. However, the landscape of eRNAs in acute myeloid leukemia (AML) and the potential roles of eRNAs in the tumor microenvironment (TME) remain unclear. Method: Gene expression data collected from The Cancer Genome Atlas (TCGA) project were combined with Histone ChIP-seq so as to reveal the comprehensive landscape of eRNAs. Single-sample gene set enrichment analysis algorithm (ssGSEA) and ESTIMATE were employed to enumerate immune cell infiltration and tumor purity. Results: Most prognostic eRNAs were enriched in immune-related pathways. Two distinct immune microenvironment patterns, the immune-active subtype and the immune-resistant subtype, were identified in AML. We further developed an eRNA-derived score (E-score) that could quantify immune microenvironment patterns and predict the response to immune checkpoint inhibitor (ICI) treatment. Finally, we established a prognostic nomogram combining E-score and other clinical features, which showed great discriminative power in both the training set [Harrell’s concordance index (C index): 0.714 (0.651–0.777), p < 0.0001] and validation set [C index: 0.684 (0.614–0.755), p < 0.0001]. Calibration of the nomogram was also validated independently. Conclusion: In this study, we systematically understood the roles of eRNAs in regulating TME diversity and complexity. Moreover, our E-score model provided the first predictive model for ICI treatment in AML.
Collapse
Affiliation(s)
- Ziming Jiang
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Eight-Year MD Program, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Junyu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaige Deng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongchang Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Miao Chen, ; Yongchang Zheng,
| | - Miao Chen
- Department of Hematology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- *Correspondence: Miao Chen, ; Yongchang Zheng,
| |
Collapse
|
15
|
Xu Q, Cao D, Fang B, Yan S, Hu Y, Guo T. Immune-related gene signature predicts clinical outcomes and immunotherapy response in acute myeloid leukemia. Cancer Med 2022; 11:3364-3380. [PMID: 35355427 PMCID: PMC9468431 DOI: 10.1002/cam4.4687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 12/05/2022] Open
Abstract
Background The immune response in the bone marrow microenvironment has implications for progression and prognosis in acute myeloid leukemia (AML). However, few immune‐related biomarkers for AML prognosis and immunotherapy response have been identified. We aimed to establish a predictive gene signature and to explore the determinants of prognosis in AML. Methods Immune‐related genes with clinical significance were screened by a weighted gene co‐expression network analysis. Seven immune‐related genes were used to establish a gene signature by a multivariate Cox regression analysis. Based on the signature, low‐ and high‐risk groups were compared with respect to the immune microenvironment, immune checkpoints, pathway activities, and mutation frequencies. The tumor immune dysfunction and exclusion (TIDE) method was used to predict the response to immune checkpoint blockade (ICB) therapy. The Connectivity Map database was used to explore small‐molecule drugs expected to treat high‐risk populations. Results A seven‐gene prognostic signature was used to classify patients into high‐ and low‐risk groups. Prognosis was poorer for patients in the former than in the latter. The high‐risk group displayed higher levels of immune checkpoint molecules (LAG3, PD‐1, CTLA4, PD‐L2, and PD‐L1), immune cell infiltration (dendritic cells, T helper 1, and gamma delta T), and somatic mutations (NPM1 and RUNX1). Moreover, hematopoietic stem cell/leukemia stem cell pathways were enriched in the high‐risk phenotype. Compared with that in the low‐risk group, the lower TIDE score for the high‐risk group implied that this group is more likely to benefit from ICB therapy. Finally, some drugs (FLT3 inhibitors and BCL inhibitors) targeting the expression profiles associated with the high‐risk group were generated using Connectivity Map. Conclusion The newly developed immune‐related gene signature is an effective biomarker for predicting prognosis in AML and provides a basis, from an immunological perspective, for the development of comprehensive therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Dedong Cao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Fang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Siqi Yan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Guo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Aru B, Soltani M, Pehlivanoglu C, Gürlü E, Ganjalikhani-Hakemi M, Yanikkaya Demirel G. Comparison of Laboratory Methods for the Clinical Follow Up of Checkpoint Blockade Therapies in Leukemia: Current Status and Challenges Ahead. Front Oncol 2022; 12:789728. [PMID: 35155232 PMCID: PMC8829140 DOI: 10.3389/fonc.2022.789728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
The development of immune checkpoint inhibitors, the monoclonal antibodies that modulate the interaction between immune checkpoint molecules or their ligands on the immune cells or tumor tissue has revolutionized cancer treatment. While there are various studies proving their efficacy in hematological malignancies, there is also a body of accumulating evidence indicating that immune checkpoint inhibitors’ clinical benefits are limited in such diseases. In addition, due to their regulatory nature that balances the immune responses, blockade of immune checkpoints may lead to toxic side effects and autoimmune responses, and even primary or acquired resistance mechanisms may restrict their success. Thus, the need for laboratory biomarkers to identify and monitor patient populations who are more likely respond to this type of therapy and the management of side effects seem critical. However, guidelines regarding the use of immune checkpoint inhibitors in hematological cancers and during follow-up are limited while there is no consensus on the laboratory parameters to be investigated for safety and efficacy of the treatment. This review aims to provide an insight into recent information on predictive and prognostic value of biomarkers and laboratory tests for the clinical follow up of hematological malignancies, with an emphasis on leukemia.
Collapse
Affiliation(s)
- Basak Aru
- Department of Immunology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Mojdeh Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Cemil Pehlivanoglu
- Department of Emergency Medicine, Hatay Training and Research Hospital, Antakya, Turkey
| | - Ege Gürlü
- Faculty of Medicine 4thYear Student, Yeditepe University, Istanbul, Turkey
| | | | | |
Collapse
|
17
|
Wen XM, Xu ZJ, Jin Y, Xia PH, Ma JC, Qian W, Lin J, Qian J. Association Analyses of TP53 Mutation With Prognosis, Tumor Mutational Burden, and Immunological Features in Acute Myeloid Leukemia. Front Immunol 2021; 12:717527. [PMID: 34745095 PMCID: PMC8566372 DOI: 10.3389/fimmu.2021.717527] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/27/2021] [Indexed: 12/28/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease related to a broad spectrum of molecular alterations. The successes of immunotherapies treating solid tumors and a deeper understanding of the immune systems of patients with hematologic malignancies have promoted the development of immunotherapies for the treatment of AML. And high tumor mutational burden (TMB) is an emerging predictive biomarker for response to immunotherapy. However, the association of gene mutation in AML with TMB and immunological features still has not been clearly elucidated. In our study, based on The Cancer Genome Atlas (TCGA) and BeatAML cohorts, 20 frequently mutated genes were found to be covered by both datasets in AML. And TP53 mutation was associated with a poor prognosis, and its mutation displayed exclusiveness with other common mutated genes in both datasets. Moreover, TP53 mutation correlated with TMB and the immune microenvironment. Gene Set Enrichment Analysis (GSEA) showed that TP53 mutation upregulated signaling pathways involved in the immune system. In summary, TP53 mutation is frequently mutated in AML, and its mutation is associated with dismal outcome, TMB, and immunological features, which may serve as a biomarker to predict immune response in AML.
Collapse
Affiliation(s)
- Xiang-Mei Wen
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ye Jin
- Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Pei-Hui Xia
- Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Qian
- Department of Otolaryngology-Head and Neck Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
18
|
Protein Arginine Methyltransferase 5 Promotes the Migration of AML Cells by Regulating the Expression of Leukocyte Immunoglobulin-Like Receptor B4. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7329072. [PMID: 34712735 PMCID: PMC8548104 DOI: 10.1155/2021/7329072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022]
Abstract
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults with poor prognosis. Especially for AML-M5 type, due to the strong cell migration ability, the possibility of extramedullary invasion is large and widespread, which leads to poor therapeutic effect. Previous studies have found that protein arginine methyltransferase 5 (PRMT5) could promote the proliferation and differentiation of leukemic cells in AML, but its regulation on the invasive ability of AML cells remains unclear. This study was designed to explore the role of PRMT5 in regulating the invasion of AML cells and to investigate the mechanisms. Patient samples were collected for detection of PRMT5 expression level. AML cells were used for exploring the function of PRMT5. The results of clinical samples showed that the expression of PRMT5 was significantly increased in newly diagnosed and recurrent AML patients, and the expression of leukocyte immunoglobulin-like receptor B4 (LILRB4) was positively correlated with the level of PRMT5. In the cell experiment in vitro, we found that when PRMT5 was knocked down, the invasion, migration, and adhesion capacities of MV-4-11 cells and THP-1 cells were decreased, and the mRNA and protein levels of LILRB4 were also decreased. Moreover, we screened related signaling pathways and found that PRMT5 affected the expression of downstream LILRB4 by activating mTOR pathway, which in turn enhanced the invasive ability of AML cells. Taken together, PRMT5 plays an important role in the invasion of AML, which acts via regulating the expression of LILRB4. PRMT5 could act as a potential therapeutic candidate for AML.
Collapse
|
19
|
Velichinskii RA, Streltsova MA, Kust SA, Sapozhnikov AM, Kovalenko EI. The Biological Role and Therapeutic Potential of NK Cells in Hematological and Solid Tumors. Int J Mol Sci 2021; 22:ijms222111385. [PMID: 34768814 PMCID: PMC8584101 DOI: 10.3390/ijms222111385] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022] Open
Abstract
NK cells are an attractive target for cancer immunotherapy due to their potent antitumor activity. The main advantage of using NK cells as cytotoxic effectors over T cells is a reduced risk of graft versus host disease. At present, several variants of NK-cell-based therapies are undergoing clinical trials and show considerable effectiveness for hematological tumors. In these types of cancers, the immune cells themselves often undergo malignant transformation, which determines the features of the disease. In contrast, the current use of NK cells as therapeutic agents for the treatment of solid tumors is much less promising. Most studies are at the stage of preclinical investigation, but few progress to clinical trials. Low efficiency of NK cell migration and functional activity in the tumor environment are currently considered the major barriers to NK cell anti-tumor therapies. Various therapeutic combinations, genetic engineering methods, alternative sources for obtaining NK cells, and other techniques are aiming at the development of promising NK cell anticancer therapies, regardless of tumorigenesis. In this review, we compare the role of NK cells in the pathogenesis of hematological and solid tumors and discuss current prospects of NK-cell-based therapy for hematological and solid tumors.
Collapse
|
20
|
Kordelas L, Buttkereit U, Heinemann FM, Horn PA, Giebel B, Beelen DW, Reinhardt HC, Rebmann V. Low Soluble Programmed Cell Death Protein 1 Levels After Allogeneic Stem Cell Transplantation Predict Moderate or Severe Chronic GvHD and Inferior Overall Survival. Front Immunol 2021; 12:694843. [PMID: 34630383 PMCID: PMC8498033 DOI: 10.3389/fimmu.2021.694843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) is an inhibitory co-receptor required for regulating immune responsiveness and maintaining immune homeostasis. As PD-1 can be released as bioactive soluble molecule, we investigated the clinical significance of soluble PD-1 (sPD-1) after allogeneic hematopoietic stem cell transplantation (HSCT) regarding graft-versus-host disease (GvHD), relapse, and overall survival (OS) in a mono-centric cohort of 82 patients. Compared to pre-HSCT and to healthy controls, post-HSCT sPD-1 plasma levels were significantly increased during an observation time of three months. Univariate analysis revealed that low sPD-1 plasma levels at month one, two or three post HSCT were associated with acute GvHD grade III-IV, the onset of moderate/severe chronic GvHD (cGvHD) and inferior OS, DFS, and TRM, respectively. No relationship was detected to relapse rates. sPD-1 plasma levels were significantly increased in ATG-treated patients compared to ATG-untreated patients. Multivariate analysis revealed that a low sPD-1 plasma levels status at one or two month(s) after HSCT is an independent indicator for inferior OS, DFS, or TRM. A low sPD-1 plasma levels status at month three post HSCT is predictive for the onset of moderate/severe cGvHD. Thus, our study pinpoints the soluble inhibitory co-receptor PD-1 as a promising candidate molecule for the prediction of clinical HSCT outcome.
Collapse
Affiliation(s)
- Lambros Kordelas
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Ulrike Buttkereit
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Falko M Heinemann
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| | - Dietrich W Beelen
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - H Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Vera Rebmann
- Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
21
|
Trial Watch: Adoptive TCR-Engineered T-Cell Immunotherapy for Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13184519. [PMID: 34572745 PMCID: PMC8469736 DOI: 10.3390/cancers13184519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/20/2021] [Accepted: 09/01/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is a type of blood cancer with an extremely grim prognosis. This is due to the fact that the majority of patients will relapse after frontline treatment. Overall survival of relapsed AML is very low, and treatment options are few. T lymphocytes harnessed with antitumor T-cell receptors (TCRs) can produce objective clinical responses in certain cancers, such as melanoma, but have not entered the main road for AML. In this review, we describe the current status of the field of TCR-T-cell therapies for AML. Abstract Despite the advent of novel therapies, acute myeloid leukemia (AML) remains associated with a grim prognosis. This is exemplified by 5-year overall survival rates not exceeding 30%. Even with frontline high-intensity chemotherapy regimens and allogeneic hematopoietic stem cell transplantation, the majority of patients with AML will relapse. For these patients, treatment options are few, and novel therapies are urgently needed. Adoptive T-cell therapies represent an attractive therapeutic avenue due to the intrinsic ability of T lymphocytes to recognize tumor cells with high specificity and efficiency. In particular, T-cell therapies focused on introducing T-cell receptors (TCRs) against tumor antigens have achieved objective clinical responses in solid tumors such as synovial sarcoma and melanoma. However, contrary to chimeric antigen receptor (CAR)-T cells with groundbreaking results in B-cell malignancies, the use of TCR-T cells for hematological malignancies is still in its infancy. In this review, we provide an overview of the status and clinical advances in adoptive TCR-T-cell therapy for the treatment of AML.
Collapse
|
22
|
Mu X, Zhang M, Wei A, Yin F, Wang Y, Hu K, Jiang J. Doxorubicin and PD-L1 siRNA co-delivery with stem cell membrane-coated polydopamine nanoparticles for the targeted chemoimmunotherapy of PCa bone metastases. NANOSCALE 2021; 13:8998-9008. [PMID: 33973580 DOI: 10.1039/d0nr08024a] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Programmed cell death ligand 1 (PD-L1) blockade has achieved great success in cancer immunotherapy. PD-L1 siRNA can restore the immune anti-tumor activity of T cells by downregulating the level of PD-L1 on tumor cells, but the efficiency of PD-1/PD-L1 monotherapy is relatively low. Doxorubicin (DOX) can induce tumor cell apoptosis, and then increase the release of tumor antigen. But the expression of PD-L1 in tumor tissues treated with DOX will be enhanced adaptively. Therefore, DOX combination with PD-L1 siRNA can produce a good synergistic anti-tumor effect. In this study, stem cell membrane (SCM) camouflaged polydopamine nanoparticles carrying DOX and PD-L1 siRNA (PDA-DOX/siPD-L1@SCM) were constructed for targeting prostate cancer (PCa) bone metastases. PDA-DOX/siPD-L1@SCM NPs could effectively enhance blood retention and improve accumulation at tumor sites. In vitro and in vivo studies demonstrated that PDA-DOX/siPD-L1@SCM NPs showed excellent performance in synergistic chemoimmunotherapy for PCa bone metastases. Hence, this study provided an effective strategy for developing biomimetic multifunctional nanoparticles for PCa bone metastasis treatment.
Collapse
Affiliation(s)
- Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, China.
| | | | | | | | | | | | | |
Collapse
|
23
|
Xie X, Hu Y, Ye T, Chen Y, Zhou L, Li F, Xi X, Wang S, He Y, Gao X, Wei W, Ma G, Li Y. Therapeutic vaccination against leukaemia via the sustained release of co-encapsulated anti-PD-1 and a leukaemia-associated antigen. Nat Biomed Eng 2021; 5:414-428. [PMID: 33046865 DOI: 10.1038/s41551-020-00624-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/03/2020] [Indexed: 02/08/2023]
Abstract
Therapeutic leukaemia vaccines have shown modest potency. Here, we show that the co-encapsulation of a leukaemia-associated epitope peptide highly expressed in leukaemia patients and of the immune checkpoint inhibitor anti-programmed-cell-death-protein-1 (anti-PD-1) in degradable poly(lactic acid) microcapsules resulted in the sustained release of the peptide and of the antibody, which led to the recruitment of activated antigen-presenting cells to the injection site, their uptake of the peptide and the transportation of the anti-PD-1 antibody to lymph nodes, enhancing the expansion of epitope-specific T cells and the activation of cytotoxic T cells. After single subcutaneous injections of vaccine formulations with different epitope peptides, mice bearing leukaemia xenografts derived from humanized cell lines or from primary cells from patients showed better therapeutic outcomes than mice receiving repeated injections of free antigen, antibody and a commercial adjuvant. The sustained release of a tumour-associated peptide and of anti-PD-1 may represent a generalizable strategy for boosting antitumour immune responses to leukaemia.
Collapse
Affiliation(s)
- Xiaoling Xie
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China.,State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Yiran Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Lijuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Xiaobo Xi
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China.,University of Chinese Academy of Sciences, Beijing, P R China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China
| | - Xiaoyong Gao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China. .,University of Chinese Academy of Sciences, Beijing, P R China.
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, P R China. .,University of Chinese Academy of Sciences, Beijing, P R China.
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, P R China. .,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, P R China.
| |
Collapse
|
24
|
Rastogi N, Baker S, Man S, Uger RA, Wong M, Coles SJ, Hodges M, Gilkes AF, Knapper S, Darley RL, Tonks A. Use of an anti-CD200-blocking antibody improves immune responses to AML in vitro and in vivo. Br J Haematol 2021; 193:155-159. [PMID: 32996123 PMCID: PMC9851282 DOI: 10.1111/bjh.17125] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 01/22/2023]
Abstract
Treatment of relapsed/resistant acute myeloid leukaemia (AML) remains a significant area of unmet patient need, the outlook for most patients remaining extremely poor. A promising approach is to augment the anti-tumour immune response in these patients; most cancers do not activate immune effector cells because they express immunosuppressive ligands. We have previously shown that CD200 (an immunosuppressive ligand) is overexpressed in AML and confers an inferior overall survival compared to CD200low/neg patients. Here we show that a fully human anti-CD200 antibody (TTI-CD200) can block the interaction of CD200 with its receptor and restore AML immune responses in vitro and in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/immunology
- Antibodies, Blocking/pharmacology
- Antigens, CD/drug effects
- Antigens, CD/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- Case-Control Studies
- Cytokine-Induced Killer Cells/immunology
- Humans
- Immunity/drug effects
- Immunity/immunology
- Immunosuppression Therapy/methods
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/therapy
- Ligands
- Mice
- Models, Animal
- Secondary Prevention/methods
- Transplantation, Heterologous/methods
Collapse
Affiliation(s)
- Namrata Rastogi
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
- School of BiosciencesEuropean Cancer Stem Cell Research InstituteCardiff UniversityCardiffCF24 4HQUK
| | - Sarah Baker
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
| | - Stephen Man
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
| | | | - Mark Wong
- Trillium Therapeutics IncMississaugaONCanada
| | - Steven J. Coles
- School of Science and the EnvironmentUniversity of WorcesterWorcestershireWR2 6AJUK
| | - Marie Hodges
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
- Cardiff Experimental and Cancer Medicine Centre (ECMC)School of MedicineCardiff UniversityCardiffCF14 4XNUK
| | - Amanda F. Gilkes
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
- Cardiff Experimental and Cancer Medicine Centre (ECMC)School of MedicineCardiff UniversityCardiffCF14 4XNUK
| | - Steven Knapper
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
| | - Richard L. Darley
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
| | - Alex Tonks
- Department of HaematologyDivision of Cancer & GeneticsSchool of MedicineCardiff UniversityCardiffCF14 4XNUK
| |
Collapse
|
25
|
Dao FT, Wang J, Yang L, Qin YZ. Development of a poor-prognostic-mutations derived immune prognostic model for acute myeloid leukemia. Sci Rep 2021; 11:4856. [PMID: 33649342 PMCID: PMC7921432 DOI: 10.1038/s41598-021-84190-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Leukemia cell-intrinsic somatic mutations and cytogenetic abnormalities have been used to define risk categories in acute myeloid leukemia (AML). In addition, since the immune microenvironment might influence prognosis and somatic mutations have been demonstrated to modulate the immune microenvironment in AML, there is need for developing and evaluating an immune prognostic model (IPM) derived from mutations associated with poor prognosis. Based on AML cases with intermediate and adverse-cytogenetic risk in the Cancer Genome Atlas (TCGA) database, 64 immune-related differentially expressed genes (DEGs) among patients with RUNX1, TP53, or ASXL1 mutations and patients without these mutations were identified. After Cox proportional hazards analysis, an IPM composed of PYCARD and PEAR1 genes was constructed. IPM defined high-risk (IPM-HR) independently predicted lower 2-year overall survival (OS) rates in both patients with intermediate and adverse-cytogenetic risks and non-M3 patients in the TCGA AML cohort. The poor prognostic impact of IPM-HR on OS was further validated by GSE71014, 37642, and 10358 downloaded from the Gene Expression Omnibus (GEO) database. Furthermore, IPM-HR was remarkably associated with higher proportions of CD8+ T cells and regulatory T cells (Tregs), lower proportions of eosinophils, and higher expression of the checkpoint molecules CTLA-4, PD-1, and LAG3 in the TCGA non-M3 AML cohort. In summary, we developed and validated an IPM derived from mutations related with poor prognosis in AML, which would provide new biomarkers for patient stratification and personalized immunotherapy.
Collapse
Affiliation(s)
- Feng-Ting Dao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Jun Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Lu Yang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China
| | - Ya-Zhen Qin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, China.
| |
Collapse
|
26
|
Tabata R, Chi S, Yuda J, Minami Y. Emerging Immunotherapy for Acute Myeloid Leukemia. Int J Mol Sci 2021; 22:1944. [PMID: 33669431 PMCID: PMC7920435 DOI: 10.3390/ijms22041944] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022] Open
Abstract
Several immune checkpoint molecules and immune targets in leukemic cells have been investigated. Recent studies have suggested the potential clinical benefits of immuno-oncology (IO) therapy against acute myeloid leukemia (AML), especially targeting CD33, CD123, and CLL-1, as well as immune checkpoint inhibitors (e.g., anti-PD (programmed cell death)-1 and anti-CTLA4 (cytotoxic T-lymphocyte-associated protein 4) antibodies) with or without conventional chemotherapy. Early-phase clinical trials of chimeric antigen receptor (CAR)-T or natural killer (NK) cells for relapsed/refractory AML showed complete remission (CR) or marked reduction of marrow blasts in a few enrolled patients. Bi-/tri-specific antibodies (e.g., bispecific T-cell engager (BiTE) and dual-affinity retargeting (DART)) exhibited 11-67% CR rates with 13-78% risk of cytokine-releasing syndrome (CRS). Conventional chemotherapy in combination with anti-PD-1/anti-CTLA4 antibody for relapsed/refractory AML showed 10-36% CR rates with 7-24 month-long median survival. The current advantages of IO therapy in the field of AML are summarized herein. However, although cancer vaccination should be included in the concept of IO therapy, it is not mentioned in this review because of the paucity of relevant evidence.
Collapse
Affiliation(s)
- Rikako Tabata
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (R.T.); (S.C.); (J.Y.)
- Department of Hematology, Kameda Medical Center, Kamogawa 296-8602, Japan
| | - SungGi Chi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (R.T.); (S.C.); (J.Y.)
| | - Junichiro Yuda
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (R.T.); (S.C.); (J.Y.)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (R.T.); (S.C.); (J.Y.)
| |
Collapse
|
27
|
Chen C, Xu L, Gao R, Wang S, Zhang Y, Wang C, Zeng C, Li Y. Transcriptome-Based Co-Expression of BRD4 and PD-1/PD-L1 Predicts Poor Overall Survival in Patients With Acute Myeloid Leukemia. Front Pharmacol 2021; 11:582955. [PMID: 33658927 PMCID: PMC7917577 DOI: 10.3389/fphar.2020.582955] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022] Open
Abstract
Positive response to PD-1/PD-L1 blockades was observed in the treatment of solid tumors. However, the clinical response to PD-1/PD-L1 blockade varied in patients with acute myeloid leukemia (AML). It is thought that there are factors other than PD-1 and PD-L1 that may affect the effect of immunotherapy. This study explored the impact of transcriptome-based co-expression of bromodomain containing 4 (BRD4) and PD-1/PD-L1 on the overall survival (OS) of patients with AML, in order to understand whether BRD4 would affect the effect of PD-1/PD-L1 blockades. Bone marrow samples from 59 AML patients in our clinical center and data of 176 patients from the Cancer Genome Atlas (TCGA) database were used for OS analysis and validation. It was found that increased expression of BRD4 was associated with poor OS in AML patients. Moreover, co-expression of BRD4 with PD-1 or PD-L1 was related to poor OS. The co-expression of BRD4 and PD-L1 was better than BRD4 and PD-1 for OS prediction. Furthermore, co-expression of BRD4 and PD-L1 was positively correlated with high tumor mutation burden, which contributed to poor OS in AML patients. Additionally, the co-expression of BRD4 and PD-L1 was associated with poor OS in non-acute promyelocytic leukemia patients with intermediate/high risk or under 60 years. Our results suggest that transcriptome-based co-expression of BRD4 and PD-L1 is a predictor for poor OS in AML patients, which might provide novel insight into designing combinational targeted therapy for AML.
Collapse
Affiliation(s)
- Cunte Chen
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Ling Xu
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China.,Department of Hematology, First Affiliated Hospital, The Clinical Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Rili Gao
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuping Zhang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Caixia Wang
- Department of Hematology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, 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
| |
Collapse
|
28
|
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: 9.5] [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
|
29
|
Ocadlikova D, Iannarone C, Redavid AR, Cavo M, Curti A. A Screening of Antineoplastic Drugs for Acute Myeloid Leukemia Reveals Contrasting Immunogenic Effects of Etoposide and Fludarabine. Int J Mol Sci 2020; 21:E6802. [PMID: 32948017 PMCID: PMC7556041 DOI: 10.3390/ijms21186802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Recent evidence demonstrated that the treatment of acute myeloid leukemia (AML) cells with daunorubicin (DNR) but not cytarabine (Ara-C) results in immunogenic cell death (ICD). In the clinical setting, chemotherapy including anthracyclines and Ara-C remains a gold standard for AML treatment. In the last decade, etoposide (Eto) and fludarabine (Flu) have been added to the standard treatment for AML to potentiate its therapeutic effect and have been tested in many trials. Very little data are available about the ability of these drugs to induce ICD. METHODS AML cells were treated with all four drugs. Calreticulin and heat shock protein 70/90 translocation, non-histone chromatin-binding protein high mobility group box 1 and adenosine triphosphate release were evaluated. The treated cells were pulsed into dendritic cells (DCs) and used for in vitro immunological tests. RESULTS Flu and Ara-C had no capacity to induce ICD-related events. Interestingly, Eto was comparable to DNR in inducing all ICD events, resulting in DC maturation. Moreover, Flu was significantly more potent in inducing suppressive T regulatory cells compared to other drugs. CONCLUSIONS Our results indicate a novel and until now poorly investigated feature of antineoplastic drugs commonly used for AML treatment, based on their different immunogenic potential.
Collapse
Affiliation(s)
- Darina Ocadlikova
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Istituto di Ematologia “Seràgnoli”, Università degli Studi, 40138 Bologna, Italy; (C.I.); (A.R.R.); (M.C.)
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| | - Clara Iannarone
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Istituto di Ematologia “Seràgnoli”, Università degli Studi, 40138 Bologna, Italy; (C.I.); (A.R.R.); (M.C.)
| | - Anna Rita Redavid
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Istituto di Ematologia “Seràgnoli”, Università degli Studi, 40138 Bologna, Italy; (C.I.); (A.R.R.); (M.C.)
| | - Michele Cavo
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Istituto di Ematologia “Seràgnoli”, Università degli Studi, 40138 Bologna, Italy; (C.I.); (A.R.R.); (M.C.)
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| | - Antonio Curti
- Azienda Ospedaliero-Universitaria di Bologna, via Albertoni 15, 40138 Bologna, Italy;
| |
Collapse
|
30
|
Zhang N, Zhang Y, Zhang P, Lou S, Chen Y, Li H, Zeng H, Shen Y, Deng J. Overexpression of annexin A5 might guide the gemtuzumab ozogamicin treatment choice in patients with pediatric acute myeloid leukemia. Ther Adv Med Oncol 2020; 12:1758835920927635. [PMID: 32636939 PMCID: PMC7310896 DOI: 10.1177/1758835920927635] [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: 10/22/2019] [Accepted: 04/27/2020] [Indexed: 12/27/2022] Open
Abstract
Background: Acute myeloid leukemia (AML) is a common hematological malignancy. Gemtuzumab
ozogamicin (GO), a humanized anti-CD33 antibody conjugated with the potent
anti-tumor antibiotic calicheamicin, represents a promising targeted therapy
for AML. Annexin A5 (ANXA5) is a proposed marker for the clinical prognosis
of AML to guide treatment choice. Methods: In total, 253 patients with pediatric AML were enrolled and divided into two
treatment groups: conventional chemotherapy alone and conventional
chemotherapy in combination with GO. Univariate, multivariate, and
Kaplan–Meier survival analyses were conducted to assess risk factors and
clinical outcomes, and to estimate hazard ratios (HRs) and their 95%
confidence interval. The level of statistical significance was set at
p < 0.05. Results: In the GO treatment group, high ANXA5 expression was
considered a favorable prognostic factor for overall survival (OS) and
event-free survival (EFS). Multivariate analysis showed that high
ANXA5 expression was an independent favorable factor
for OS (HR = 0.629, p = 0.084) and EFS (HR = 0.544,
p = 0.024) distinct from the curative effect of GO
treatment. When all patients were again divided into two groups, this time
based on the median expression of ANXA5, patients
undergoing chemotherapy combined with GO had significantly better OS
(p = 0.0012) and EFS (p = 0.0003) in
the ANXA5 high-expression group. Gene set enrichment
analysis identified a relevant series of pathways associated with
glutathione metabolism, leukocyte transendothelial migration, and
hematopoietic cell lineage. Conclusion: The expression level of ANXA5 can help optimize the
treatment regimen for individual patients, and patients with overexpression
of ANXA5 may circumvent poor outcomes from chemotherapy
combined with GO.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Ying Zhang
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Ping Zhang
- Hematology Laboratory, The Second Affiliated Hospital, Chongqing Medical University, Yuzhong, Chongqing, P.R. China
| | - Shifeng Lou
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Ying Chen
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Huan Li
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Hanqing Zeng
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Yan Shen
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, Jiangnan, Chongqing, P.R. China
| | - Jianchuan Deng
- Department of Hematology, The Second Affiliated Hospital, Chongqing Medical University, 76 Linjiang Road, Chongqing, 400010, P.R. China
| |
Collapse
|
31
|
van Ens D, Mousset CM, Hutten TJA, van der Waart AB, Campillo-Davo D, van der Heijden S, Vodegel D, Fredrix H, Woestenenk R, Parga-Vidal L, Jansen JH, Schaap NPM, Lion E, Dolstra H, Hobo W. PD-L1 siRNA-mediated silencing in acute myeloid leukemia enhances anti-leukemic T cell reactivity. Bone Marrow Transplant 2020; 55:2308-2318. [PMID: 32528120 DOI: 10.1038/s41409-020-0966-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/26/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) is an immune-susceptible malignancy, as demonstrated by its responsiveness to allogeneic stem cell transplantation (alloSCT). However, by employing inhibitory signaling pathways, including PD-1/PD-L1, leukemia cells suppress T cell-mediated immune attack. Notably, impressive clinical efficacy has been obtained with PD-1/PD-L1 blocking antibodies in cancer patients. Yet, these systemic treatments are often accompanied by severe toxicity, especially after alloSCT. Here, we investigated RNA interference technology as an alternative strategy to locally interfere with PD-1/PD-L1 signaling in AML. We demonstrated efficient siRNA-mediated PD-L1 silencing in HL-60 and patients' AML cells. Importantly, WT1-antigen T cell receptor+ PD-1+ 2D3 cells showed increased activation toward PD-L1 silenced WT1+ AML. Moreover, PD-L1 silenced AML cells significantly enhanced the activation, degranulation, and IFN-γ production of minor histocompatibility antigen-specific CD8+ T cells. Notably, PD-L1 silencing was equally effective as PD-1 antibody blockade. Together, our study demonstrates that PD-L1 silencing may be an effective strategy to augment AML immune-susceptibility. This provides rationale for further development of targeted approaches to locally interfere with immune escape mechanisms in AML, thereby minimizing severe toxicity. In combination with alloSCT and/or adoptive T cell transfer, this strategy could be very appealing to boost graft-versus-leukemia immunity and improve outcome in AML patients.
Collapse
Affiliation(s)
- Diede van Ens
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Charlotte M Mousset
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tim J A Hutten
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Anniek B van der Waart
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Sanne van der Heijden
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Denise Vodegel
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hanny Fredrix
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rob Woestenenk
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Loreto Parga-Vidal
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joop H Jansen
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nicolaas P M Schaap
- Department of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute, Faculty of Medicine & Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Harry Dolstra
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willemijn Hobo
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
32
|
Sivori S, Meazza R, Quintarelli C, Carlomagno S, Della Chiesa M, Falco M, Moretta L, Locatelli F, Pende D. NK Cell-Based Immunotherapy for Hematological Malignancies. J Clin Med 2019; 8:E1702. [PMID: 31623224 PMCID: PMC6832127 DOI: 10.3390/jcm8101702] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) lymphocytes are an integral component of the innate immune system and represent important effector cells in cancer immunotherapy, particularly in the control of hematological malignancies. Refined knowledge of NK cellular and molecular biology has fueled the interest in NK cell-based antitumor therapies, and recent efforts have been made to exploit the high potential of these cells in clinical practice. Infusion of high numbers of mature NK cells through the novel graft manipulation based on the selective depletion of T cells and CD19+ B cells has resulted into an improved outcome in children with acute leukemia given human leucocyte antigen (HLA)-haploidentical hematopoietic transplantation. Likewise, adoptive transfer of purified third-party NK cells showed promising results in patients with myeloid malignancies. Strategies based on the use of cytokines or monoclonal antibodies able to induce and optimize NK cell activation, persistence, and expansion also represent a novel field of investigation with remarkable perspectives of favorably impacting on outcome of patients with hematological neoplasia. In addition, preliminary results suggest that engineering of mature NK cells through chimeric antigen receptor (CAR) constructs deserve further investigation, with the goal of obtaining an "off-the-shelf" NK cell bank that may serve many different recipients for granting an efficient antileukemia activity.
Collapse
Affiliation(s)
- Simona Sivori
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy (S.C.); (M.D.C.)
- Centre of Excellence for Biomedical Research, University of Genoa, 16132 Genoa, Italy
| | - Raffaella Meazza
- Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| | - Concetta Quintarelli
- Department of Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy; (C.Q.); (F.L.)
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy (S.C.); (M.D.C.)
| | - Mariella Della Chiesa
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy (S.C.); (M.D.C.)
- Centre of Excellence for Biomedical Research, University of Genoa, 16132 Genoa, Italy
| | - Michela Falco
- Integrated Department of Services and Laboratories, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Ospedale Pediatrico Bambino Gesù, 00146 Rome, Italy;
| | - Franco Locatelli
- Department of Hematology/Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, 00165 Rome, Italy; (C.Q.); (F.L.)
- Department of Gynecology/Obstetrics and Pediatrics, Sapienza University, 00185 Rome, Italy
| | - Daniela Pende
- Department of Integrated Oncological Therapies, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy;
| |
Collapse
|
33
|
Ocadlikova D, Lecciso M, Isidori A, Loscocco F, Visani G, Amadori S, Cavo M, Curti A. Chemotherapy-Induced Tumor Cell Death at the Crossroads Between Immunogenicity and Immunotolerance: Focus on Acute Myeloid Leukemia. Front Oncol 2019; 9:1004. [PMID: 31649875 PMCID: PMC6794495 DOI: 10.3389/fonc.2019.01004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/18/2019] [Indexed: 01/25/2023] Open
Abstract
In solid tumors and hematological malignancies, including acute myeloid leukemia, some chemotherapeutic agents, such as anthracyclines, have proven to activate an immune response via dendritic cell-based cross-priming of anti-tumor T lymphocytes. This process, known as immunogenic cell death, is characterized by a variety of tumor cell modifications, i.e., cell surface translocation of calreticulin, extracellular release of adenosine triphosphate and pro-inflammatory factors, such as high mobility group box 1 proteins. However, in addition to with immunogenic cell death, chemotherapy is known to induce inflammatory modifications within the tumor microenvironment, which may also elicit immunosuppressive pathways. In particular, DCs may be driven to acquire tolerogenic features, such as the overexpression of indoleamine 2,3-dioxygensase 1, which may ultimately hamper anti-tumor T-cells via the induction of T regulatory cells. The aim of this review is to summarize the current knowledge about the mechanisms and effects by which chemotherapy results in both activation and suppression of anti-tumor immune response. Indeed, a better understanding of the whole process underlying chemotherapy-induced alterations of the immunological tumor microenvironment has important clinical implications to fully exploit the immunogenic potential of anti-leukemia agents and tune their application.
Collapse
Affiliation(s)
- Darina Ocadlikova
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| | - Mariangela Lecciso
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| | - Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Sergio Amadori
- Department of Medicine, Institute of Hematology, University Hospital Tor Vergata, Rome, Italy
| | - Michele Cavo
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| | - Antonio Curti
- Department of Hematology and Oncology, University Hospital S.Orsola-Malpighi, Institute of Hematology "L. and A. Seràgnoli", Bologna, Italy
| |
Collapse
|
34
|
Hansrivijit P, Gale RP, Barrett J, Ciurea SO. Cellular therapy for acute myeloid Leukemia – Current status and future prospects. Blood Rev 2019; 37:100578. [DOI: 10.1016/j.blre.2019.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/23/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
|
35
|
Natural Killer Immunotherapy for Minimal Residual Disease Eradication Following Allogeneic Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia. Int J Mol Sci 2019; 20:ijms20092057. [PMID: 31027331 PMCID: PMC6539946 DOI: 10.3390/ijms20092057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 12/17/2022] Open
Abstract
The most common cause of death in patients with acute myeloid leukemia (AML) who receive allogeneic hematopoietic stem cell transplantation (allo-HSCT) is AML relapse. Therefore, additive therapies post allo-HSCT have significant potential to prevent relapse. Natural killer (NK)-cell-based immunotherapies can be incorporated into the therapeutic armamentarium for the eradication of AML cells post allo-HSCT. In recent studies, NK cell-based immunotherapies, the use of adoptive NK cells, NK cells in combination with cytokines, immune checkpoint inhibitors, bispecific and trispecific killer cell engagers, and chimeric antigen receptor-engineered NK cells have all shown antitumor activity in AML patients. In this review, we will discuss the current strategies with these NK cell-based immunotherapies as possible therapies to cure AML patients post allo-HSCT. Additionally, we will discuss various means of immune escape in order to further understand the mechanism of NK cell-based immunotherapies against AML.
Collapse
|
36
|
Toffalori C, Zito L, Gambacorta V, Riba M, Oliveira G, Bucci G, Barcella M, Spinelli O, Greco R, Crucitti L, Cieri N, Noviello M, Manfredi F, Montaldo E, Ostuni R, Naldini MM, Gentner B, Waterhouse M, Zeiser R, Finke J, Hanoun M, Beelen DW, Gojo I, Luznik L, Onozawa M, Teshima T, Devillier R, Blaise D, Halkes CJM, Griffioen M, Carrabba MG, Bernardi M, Peccatori J, Barlassina C, Stupka E, Lazarevic D, Tonon G, Rambaldi A, Cittaro D, Bonini C, Fleischhauer K, Ciceri F, Vago L. Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation. Nat Med 2019; 25:603-611. [PMID: 30911134 DOI: 10.1038/s41591-019-0400-z] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 02/15/2019] [Indexed: 01/17/2023]
Abstract
Transplantation of hematopoietic cells from a healthy individual (allogeneic hematopoietic cell transplantation (allo-HCT)) demonstrates that adoptive immunotherapy can cure blood cancers: still, post-transplantation relapses remain frequent. To explain their drivers, we analyzed the genomic and gene expression profiles of acute myeloid leukemia (AML) blasts purified from patients at serial time-points during their disease history. We identified a transcriptional signature specific for post-transplantation relapses and highly enriched in immune-related processes, including T cell costimulation and antigen presentation. In two independent patient cohorts we confirmed the deregulation of multiple costimulatory ligands on AML blasts at post-transplantation relapse (PD-L1, B7-H3, CD80, PVRL2), mirrored by concomitant changes in circulating donor T cells. Likewise, we documented the frequent loss of surface expression of HLA-DR, -DQ and -DP on leukemia cells, due to downregulation of the HLA class II regulator CIITA. We show that loss of HLA class II expression and upregulation of inhibitory checkpoint molecules represent alternative modalities to abolish AML recognition from donor-derived T cells, and can be counteracted by interferon-γ or checkpoint blockade, respectively. Our results demonstrate that the deregulation of pathways involved in T cell-mediated allorecognition is a distinctive feature and driver of AML relapses after allo-HCT, which can be rapidly translated into personalized therapies.
Collapse
Affiliation(s)
- Cristina Toffalori
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Laura Zito
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Valentina Gambacorta
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Michela Riba
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giacomo Oliveira
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gabriele Bucci
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Matteo Barcella
- Genomic and Bioinformatics Unit, Department of Health Sciences, University of Milano, Milano, Italy
| | - Orietta Spinelli
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Lara Crucitti
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy.,University of Milano, Milano, Italy
| | - Nicoletta Cieri
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy.,University of Milano, Milano, Italy
| | - Maddalena Noviello
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Francesco Manfredi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Elisa Montaldo
- Genomics of the Innate Immune System Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Renato Ostuni
- Genomics of the Innate Immune System Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Matteo M Naldini
- Translational Stem Cell and Leukemia Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Bernhard Gentner
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Translational Stem Cell and Leukemia Unit, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Miguel Waterhouse
- Department of Hematology, Oncology and Stem Cell Transplantation, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology, Oncology and Stem Cell Transplantation, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Jurgen Finke
- Department of Hematology, Oncology and Stem Cell Transplantation, Universitätsklinikum Freiburg, Freiburg, Germany
| | - Maher Hanoun
- Department of Bone Marrow Transplantation, Universitätsklinikum Essen, Essen, Germany
| | - Dietrich W Beelen
- Department of Bone Marrow Transplantation, Universitätsklinikum Essen, Essen, Germany
| | - Ivana Gojo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leo Luznik
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Masahiro Onozawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Sapporo, Japan
| | - Raynier Devillier
- Department of Haematology, Institut Paoli Calmettes, Marseille, France
| | - Didier Blaise
- Department of Haematology, Institut Paoli Calmettes, Marseille, France
| | | | - Marieke Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matteo G Carrabba
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Massimo Bernardi
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Jacopo Peccatori
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Cristina Barlassina
- Genomic and Bioinformatics Unit, Department of Health Sciences, University of Milano, Milano, Italy
| | - Elia Stupka
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dejan Lazarevic
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giovanni Tonon
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy.,Department of Oncology and Hemato-Oncology, University of Milano, Milano, Italy
| | - Davide Cittaro
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,San Raffaele Vita-Salute University, Milano, Italy
| | - Katharina Fleischhauer
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy.,Institute for Experimental Cellular Therapy, Universitätsklinikum Essen, Essen, Germany
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy.,San Raffaele Vita-Salute University, Milano, Italy
| | - Luca Vago
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, Division of Immunology, Transplantation and Infectious Disease, IRCCS San Raffaele Scientific Institute, Milano, Italy. .,Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milano, Italy.
| |
Collapse
|
37
|
Hattori N, Kawaguchi Y, Sasaki Y, Shimada S, Murai S, Abe M, Baba Y, Watanuki M, Fujiwara S, Arai N, Kabasawa N, Tsukamoto H, Uto Y, Yanagisawa K, Saito B, Harada H, Nakamaki T. Monitoring TIGIT/DNAM-1 and PVR/PVRL2 Immune Checkpoint Expression Levels in Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia. Biol Blood Marrow Transplant 2019; 25:861-867. [PMID: 30639819 DOI: 10.1016/j.bbmt.2019.01.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/03/2019] [Indexed: 12/19/2022]
Abstract
After allogeneic stem cell transplantation (alloSCT), several immune checkpoints play an important role in the antileukemic immune response in the bone marrow (BM) microenvironment. However, immune checkpoint expression levels in the BM have not been reported after alloSCT in patients with acute myeloid leukemia (AML). We investigated the clinical impact of immune checkpoint expression in BM samples after alloSCT for AML. Higher expression of T cell immunoreceptor with Ig and ITIM domains (TIGIT) was associated with a decreased incidence of acute graft-versus-host disease (P = .048) and poor overall (P = .046) and progression-free survival (P = 0.024). In addition, higher expression of TIGIT at engraftment after alloSCT was correlated with a decreased number of natural killer cells in BM (P = .019). Monitoring TIGIT expression in the BM could be useful for predicting outcome after alloSCT for AML. Our findings raise the possibility that blockade of TIGIT would improve survival.
Collapse
Affiliation(s)
- Norimichi Hattori
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan.
| | - Yukiko Kawaguchi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yohei Sasaki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shotaro Shimada
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - So Murai
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Maasa Abe
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yuta Baba
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Megumi Watanuki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Shun Fujiwara
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Nana Arai
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Nobuyuki Kabasawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroyuki Tsukamoto
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yui Uto
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Kouji Yanagisawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Bungo Saito
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hiroshi Harada
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Nakamaki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
38
|
Mechanisms of immune escape after allogeneic hematopoietic cell transplantation. Blood 2018; 133:1290-1297. [PMID: 30578254 DOI: 10.1182/blood-2018-10-846824] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/15/2018] [Indexed: 02/04/2023] Open
Abstract
Relapse of the original disease is a major cause of death after allogeneic hematopoietic cell transplantation for acute leukemias. There is growing evidence that relapses may be explained not only by resistance to chemotherapy but also by the escape of tumor cells from the control of the allogeneic immune response. Mechanisms of immune evasion can involve abrogation of leukemia cell recognition due to loss of HLA genes, immunosuppression by immune-checkpoint ligand expression, production of anti-inflammatory factors, release of metabolically active enzymes, loss of proinflammatory cytokine production, and acquisition of novel driver mutations that promote leukemia outgrowth. These mechanisms, and therapeutic targeting of immune escape, will be discussed. We divide the evidence in support of immune-escape mechanisms into animal studies, human laboratory studies, and human clinical experience. A better understanding of the molecular pathways connected to immune escape and relapse may help to improve our therapeutic armamentarium against acute myeloid leukemia relapse.
Collapse
|
39
|
Bifunctional PD-1 × αCD3 × αCD33 fusion protein reverses adaptive immune escape in acute myeloid leukemia. Blood 2018; 132:2484-2494. [PMID: 30275109 DOI: 10.1182/blood-2018-05-849802] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022] Open
Abstract
The CD33-targeting bispecific T-cell engager (BiTE) AMG 330 proved to be highly efficient in mediating cytolysis of acute myeloid leukemia (AML) cells in vitro and in mouse models. Yet, T-cell activation is correlated with upregulation of programmed cell death-ligand 1 (PD-L1) and other inhibitory checkpoints on AML cells that confer adaptive immune resistance. PD-1 and PD-L1 blocking agents may counteract T-cell dysfunction, however, at the expense of broadly distributed immune-related adverse events (irAEs). We developed a bifunctional checkpoint inhibitory T cell-engaging (CiTE) antibody that combines T-cell redirection to CD33 on AML cells with locally restricted immune checkpoint blockade. This is accomplished by fusing the extracellular domain of PD-1 (PD-1ex), which naturally holds a low affinity to PD-L1, to an αCD3.αCD33 BiTE-like scaffold. By a synergistic effect of checkpoint blockade and avidity-dependent binding, the PD-1ex attachment increases T-cell activation (3.3-fold elevation of interferon-γ) and leads to efficient and highly selective cytotoxicity against CD33+PD-L1+ cell lines (50% effective concentration = 2.3-26.9 pM) as well as patient-derived AML cells (n = 8). In a murine xenograft model, the CiTE induces complete AML eradication without initial signs of irAEs as measured by body weight loss. We conclude that our molecule preferentially targets AML cells, whereas high-affinity blockers, such as clinically approved anticancer agents, also address PD-L1+ non-AML cells. By combining the high efficacy of T-cell engagers with immune checkpoint blockade in a single molecule, we expect to minimize irAEs associated with the systemic application of immune checkpoint inhibitors and suggest high therapeutic potential, particularly for patients with relapsed/ refractory AML.
Collapse
|
40
|
Germline mutations in the bone marrow microenvironment and dysregulated hematopoiesis. Exp Hematol 2018; 66:17-26. [PMID: 30076950 DOI: 10.1016/j.exphem.2018.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
The relationship between the hematopoietic stem cell (HSC) population and its surrounding bone marrow microenvironment is a rapidly evolving area of research. Normal HSC processes rely heavily on a complex communication network involving various marrow niches. Although leukemogenesis largely results from abnormal genetic activity within the leukemia stem cell itself, mounting evidence indicates a significant contributory role played by marrow niche dysregulation. Furthermore, numerous instances of activating or inactivating germline mutations within marrow microenvironment cells have been shown to be sufficient for development of myelodysplastic syndrome, myeloproliferative neoplasm, and acute myeloid leukemia, even in the context of wild-type HSCs. Recent evidence suggests that targeting aberrant chemokine production from germline-mutated marrow stromal cells can potentially reverse the process of leukemogenesis. This elaborate interplay between the HSC population and the marrow microenvironment allows for a number of unique clinical possibilities in efforts to induce remission, enhance chemosensitivity, manage relapsed disease, and prevent leukemia development, both in de novo and germline mutation-associated leukemias, including the use of targeted cytokine/chemokine inhibitors, immune checkpoint blockade, CXCR4/CXCL12 axis antagonists, and combined allogeneic HSC and mesenchymal stem cell transplantation. In this review, we discuss the pathways underlying normal and abnormal bone marrow niche functioning, the relationship between germline mutations in the stem cell microenvironment and dysregulated hematopoiesis, and future clinical perspectives that may be particularly applicable to prevention and treatment of germline-associated leukemias.
Collapse
|
41
|
Wang Z, Huang W, Cen B, Wei Y, Liao L, Li G, Ji A. [Small interfering RNA-mediated programmed cell death-ligand 1 silencing in human glioma cells enhances human CD8 + T lymphocyte cytotoxicity in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:800-806. [PMID: 33168513 DOI: 10.3969/j.issn.1673-4254.2018.07.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the effect of small interfering RNA (siRNA)-mediated silencing of programmed cell deathligand 1 (PD-L1) in human glioma cells on the cytotoxicity of human CD8+T lymphocytes against the modified tumor cells. METHODS A siRNA sequence targeting PD-L1 gene was designed and transfected into human glioma U87 MG cells via lipofectamine 2000, and the gene silencing effect was validated using RT-qPCR, Western blotting, and flow cytometry. The transfected cells were co-cultured with human CD8+T lymphocytes, and the apoptosis of the tumor cells was analyzed with flow cytometry. RESULTS The siRNA sequence showed strong PD-L1 gene-silencing effect at both mRNA and protein levels in U87 MG cells. Compared with the control cells, the transfected U87 MG cells showed significantly increased vulnerability to the cytotoxicity of human CD8+T cells and an obvious reduction of proliferative activity in the co-culture (P < 0.05). CONCLUSIONS Transfection of human glioma U87 MG cells with the specific siRNA targeting PD-L1 obviously enhances the toxicity of human T lymphocytes in the co-culture.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China.,R&D Center, Nanjing Pharmaceutical Factory Co., Ltd., Nanjing 210007, China.,Department of Pharmacy, Shaoyang Central Hospital, Shaoyang 422000, China
| | - Wen Huang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Bohong Cen
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Yuanyi Wei
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Lumin Liao
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Guoxian Li
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Aimin Ji
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China.,R&D Center, Nanjing Pharmaceutical Factory Co., Ltd., Nanjing 210007, China.,Department of Radiation Oncology, Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 510282, China
| |
Collapse
|