1
|
Tan X, Zhao X. B7-H3 in acute myeloid leukemia: From prognostic biomarker to immunotherapeutic target. Chin Med J (Engl) 2024; 137:2540-2551. [PMID: 38595093 DOI: 10.1097/cm9.0000000000003099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Indexed: 04/11/2024] Open
Abstract
ABSTRACT B7-H3 (CD276), an immune checkpoint protein of the B7 family, exhibits significant upregulation in solid tumors and hematologic malignancies, exerting a crucial role in their pathophysiology. The distinct differential expression of B7-H3 between tumors and normal tissues and its multifaceted involvement in tumor pathogenesis position it as a promising therapeutic target for tumors. In the context of acute myeloid leukemia (AML), B7-H3 is prominently overexpressed and closely associated with unfavorable prognoses, yet it has remained understudied. Despite various ongoing clinical trials demonstrating the potential efficacy of immunotherapies targeting B7-H3, the precise underlying mechanisms responsible for B7-H3-mediated proliferation and immune evasion in AML remain enigmatic. In view of this, we comprehensively outline the current research progress concerning B7-H3 in AML, encompassing in-depth discussions on its structural attributes, receptor interactions, expression profiles, and biological significance in normal tissues and AML. Moreover, we delve into the protumor effects of B7-H3 in AML, examine the intricate mechanisms that underlie its function, and discuss the emerging application of B7-H3-targeted therapy in AML treatment. By juxtaposing B7-H3 with other molecules within the B7 family, this review emphasizes the distinctive advantages of B7-H3, not only as a valuable prognostic biomarker but also as a highly promising immunotherapeutic target in AML.
Collapse
Affiliation(s)
- Xiao Tan
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing 100044, China
| | | |
Collapse
|
2
|
Musil J, Ptacek A, Vanikova S. OMIP-106: A 30-color panel for analysis of check-point inhibitory networks in the bone marrow of acute myeloid leukemia patients. Cytometry A 2024; 105:729-736. [PMID: 39192598 DOI: 10.1002/cyto.a.24892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 08/29/2024]
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia diagnosed in adults. Despite advances in medical care, the treatment of AML still faces many challenges, such as treatment-related toxicities, that limit the use of high-intensity chemotherapy, especially in elderly patients. Currently, various immunotherapeutic approaches, that is, CAR-T cells, BiTEs, and immune checkpoint inhibitors, are being tested in clinical trials to prolong remission and improve the overall survival of AML patients. However, early reports show only limited benefits of these interventions and only in a subset of patients, showing the need for better patient stratification based on immunological markers. We have therefore developed and optimized a 30-color panel for evaluation of effector immune cell (NK cells, γδ T cells, NKT-like T cells, and classical T cells) infiltration into the bone marrow and analysis of their phenotype with regard to their differentiation, expression of inhibitory (PD-1, TIGIT, Tim3, NKG2A) and activating receptors (DNAM-1, NKG2D). We also evaluate the immune evasive phenotype of CD33+ myeloid cells, CD34+CD38-, and CD34+CD38+ hematopoietic stem and progenitor cells by analyzing the expression of inhibitory ligands such as PD-L1, CD112, CD155, and CD200. Our panel can be a valuable tool for patient stratification in clinical trials and can also be used to broaden our understanding of check-point inhibitory networks in AML.
Collapse
Affiliation(s)
- Jan Musil
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Antonin Ptacek
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University Prague, Prague, Czech Republic
| | - Sarka Vanikova
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University Prague, Prague, Czech Republic
| |
Collapse
|
3
|
Stefańczyk SA, Hayn C, Heitmann J, Jung S, Zekri L, Märklin M. Expression and Prognostic Value of a Novel B7-H3 (CD276) Antibody in Acute Myeloid Leukemia. Cancers (Basel) 2024; 16:2455. [PMID: 39001517 PMCID: PMC11240323 DOI: 10.3390/cancers16132455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Despite recent advances in immunophenotyping, the prognosis of acute myeloid leukemia (AML) is still mainly estimated using age and genetic markers. As the genetic heterogeneity of AML patients is high, flow cytometry-based classification with appropriate biomarkers can efficiently complement risk stratification and treatment selection. An increased expression of B7-H3 (CD276), an immune checkpoint protein, has been reported and associated with poor prognosis. However, the available data are limited and heterogeneous. Here, we used a novel, proprietary murine anti-B7-H3 8H8 antibody for the flow cytometric analysis of B7-H3 expression in AML blasts from 77 patients. Our antibody reliably detected substantial B7-H3 expression in 62.3% of AML patients. B7-H3 expression was higher in the monocytic French-American-British (FAB) M5 group and in intermediate and poor risk patients according to the European Leukemia Network. Using receiver operating characteristics (ROCs), we identified a specific fluorescence intensity cut-off of 4.45 to discriminate between B7-H3high and B7-H3low expression. High B7-H3 expression was associated with shorter overall survival (OS) and progression-free survival (PFS). In conclusion, we have developed a novel B7-H3 antibody that serves as a new tool for the detection of B7-H3 expression in AML and may help to facilitate risk stratification and treatment selection in AML patients.
Collapse
Affiliation(s)
- Sylwia A. Stefańczyk
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Clara Hayn
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Jonas Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Susanne Jung
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
- Department of Peptide−based Immunotherapy, Institute of Immunology, University and University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Latifa Zekri
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, 72076 Tuebingen, Germany; (S.A.S.); (C.H.); (J.H.); (S.J.)
- Cluster of Excellence iFIT (EXC 2180) ‘Image-Guided and Functionally Instructed Tumor Therapies’, Eberhard Karls University Tuebingen, 72076 Tuebingen, 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
|
Garg S, Ni W, Griffin JD, Sattler M. Chimeric Antigen Receptor T Cell Therapy in Acute Myeloid Leukemia: Trials and Tribulations. Hematol Rep 2023; 15:608-626. [PMID: 37987319 PMCID: PMC10660693 DOI: 10.3390/hematolrep15040063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that is often associated with relapse and drug resistance after standard chemotherapy or targeted therapy, particularly in older patients. Hematopoietic stem cell transplants are looked upon as the ultimate salvage option with curative intent. Adoptive cell therapy using chimeric antigen receptors (CAR) has shown promise in B cell malignancies and is now being investigated in AML. Initial clinical trials have been disappointing in AML, and we review current strategies to improve efficacy for CAR approaches. The extensive number of clinical trials targeting different antigens likely reflects the genetic heterogeneity of AML. The limited number of patients reported in multiple early clinical studies makes it difficult to draw conclusions about CAR safety, but it does suggest that the efficacy of this approach in AML lags behind the success observed in B cell malignancies. There is a clear need not only to improve CAR design but also to identify targets in AML that show limited expression in normal myeloid lineage cells.
Collapse
Affiliation(s)
- Swati Garg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Ni
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - James D. Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Bolkun L, Tynecka M, Walewska A, Bernatowicz M, Piszcz J, Cichocka E, Wandtke T, Czemerska M, Wierzbowska A, Moniuszko M, Grubczak K, Eljaszewicz A. The Association between Immune Checkpoint Proteins and Therapy Outcomes in Acute Myeloid Leukaemia Patients. Cancers (Basel) 2023; 15:4487. [PMID: 37760457 PMCID: PMC10526931 DOI: 10.3390/cancers15184487] [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: 08/07/2023] [Revised: 08/29/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The development of novel drugs with different mechanisms of action has dramatically changed the treatment landscape of AML patients in recent years. Considering a significant dysregulation of the immune system, inhibitors of immune checkpoint (ICI) proteins provide a substantial therapeutic option for those subjects. However, use of ICI in haematological malignancies remains very limited, in contrast to their wide use in solid tumours. Here, we analysed expression patterns of the most promising selected checkpoint-based therapeutic targets in AML patients. Peripheral blood of 72 untreated AML patients was used for flow cytometric analysis. Expression of PD-1, PD-L1, CTLA-4, and B7-H3 was assessed within CD4+ (Th) lymphocytes and CD33+ blast cells. Patients were stratified based on therapy outcome and cytogenetic molecular risk. AML non-responders (NR) showed a higher frequency of PD-1 in Th cells compared to those with complete remission (CR). Reduced blast cell level of CTLA-4 was another factor differentiating CR from NR subjects. Elevated levels of PD-1 were associated with a trend for poorer patients' survival. Additionally, prognosis for AML patients was worse in case of a higher frequency of B7-H3 in Th lymphocytes. In summary, we showed the significance of selected ICI as outcome predictors in AML management. Further, multicentre studies are required for validation of those data.
Collapse
Affiliation(s)
- Lukasz Bolkun
- Department of Haematology, Medical University of Bialystok, 15-276 Bialystok, Poland (J.P.)
| | - Marlena Tynecka
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland (A.W.); (M.M.)
| | - Alicja Walewska
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland (A.W.); (M.M.)
| | - Malgorzata Bernatowicz
- Department of Haematology, Medical University of Bialystok, 15-276 Bialystok, Poland (J.P.)
| | - Jaroslaw Piszcz
- Department of Haematology, Medical University of Bialystok, 15-276 Bialystok, Poland (J.P.)
| | - Edyta Cichocka
- Department of Haematology, Rydygiera Hospital in Torun, 87-100 Torun, Poland;
| | - Tomasz Wandtke
- Department of Lung Diseases, Neoplasms and Tuberculosis, Nicolaus Copernicus University in Torun, 85-326 Bydgoszcz, Poland;
| | - Magdalena Czemerska
- Department of Hematology, Medical University of Lodz, 93-510 Lodz, Poland (A.W.)
| | | | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland (A.W.); (M.M.)
- Department of Allergology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Kamil Grubczak
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland (A.W.); (M.M.)
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland (A.W.); (M.M.)
- Tissue and Cell Bank, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
7
|
Guo X, Yu S, Ren X, Li L. Immune checkpoints represent a promising breakthrough in targeted therapy and prognosis of myelodysplastic syndrome. Heliyon 2023; 9:e19222. [PMID: 37810157 PMCID: PMC10558320 DOI: 10.1016/j.heliyon.2023.e19222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 06/27/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a hematological malignancy of undetermined etiology, possibly linked to chromosomal structural alterations, genetic mutations, presentation and carcinogenicity of variant antigens on cell surface, and the generation of pro-inflammatory microenvironment in the bone marrow. Current drugs are unable to cure this disease, and therefore, decreasing the survival and proliferation of malignant cells to delay disease progression and extend the survival time of patients becomes the primary approach to management. In recent years, the immune system has received increasing attention for its potential role in the occurrence and development of MDS, leading to the emergence of immunoregulation as a viable treatment option. The current review provides a brief overview of pathogenesis of MDS and current treatment principles. In the meantime, the significance of immune proteins in treatment and prognosis of MDS is also discussed.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Shunjie Yu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Xiaotong Ren
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, Heping District 154 Anshan Road, Tianjin, China
| |
Collapse
|
8
|
Kamali AN, Bautista JM, Eisenhut M, Hamedifar H. Immune checkpoints and cancer immunotherapies: insights into newly potential receptors and ligands. Ther Adv Vaccines Immunother 2023; 11:25151355231192043. [PMID: 37662491 PMCID: PMC10469281 DOI: 10.1177/25151355231192043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 07/14/2023] [Indexed: 09/05/2023] Open
Abstract
Checkpoint markers and immune checkpoint inhibitors have been increasingly identified and developed as potential immunotherapeutic targets in various human cancers. Despite valuable efforts to discover novel immune checkpoints and their ligands, the precise roles of their therapeutic functions, as well as the broad identification of their counterpart receptors, remain to be addressed. In this context, it has been suggested that various putative checkpoint receptors can be induced upon activation. In the tumor microenvironment, T cells, as crucial immune response against malignant diseases as well as other immune central effector cells, such as natural killer cells, are regulated via co-stimulatory or co-inhibitory signals from immune or tumor cells. Studies have shown that exposure of T cells to tumor antigens upregulates the expression of inhibitory checkpoint receptors, leading to T-cell dysfunction or exhaustion. Although targeting immune checkpoint regulators has shown relative clinical efficacy in some tumor types, most trials in the field of cancer immunotherapies have revealed unsatisfactory results due to de novo or adaptive resistance in cancer patients. To overcome these obstacles, combinational therapies with newly discovered inhibitory molecules or combined blockage of several checkpoints provide a rationale for further research. Moreover, precise identification of their receptors counterparts at crucial checkpoints is likely to promise effective therapies. In this review, we examine the prospects for the application of newly emerging checkpoints, such as T-cell immunoglobulin and mucin domain 3, lymphocyte activation gene-3, T-cell immunoreceptor with Ig and ITIM domains (TIGIT), V-domain Ig suppressor of T-cell activation (VISTA), new B7 family proteins, and B- and T-cell lymphocyte attenuator, in association with immunotherapy of malignancies. In addition, their clinical and biological significance is discussed, including their expression in various human cancers, along with their roles in T-cell-mediated immune responses.
Collapse
Affiliation(s)
- Ali N. Kamali
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Simin Dasht Industrial Area, Karaj, Iran
- CinnaGen Research and Production Co., Alborz 3165933155, Iran
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Sciences, Complutense University of Madrid, Madrid, Spain
- Research Institute Hospital 12 de Octubre, Madrid, Spain
| | - Michael Eisenhut
- Department of Pediatrics, Luton and Dunstable University Hospital NHS Foundation Trust, Luton, UK
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| |
Collapse
|
9
|
Villar S, Ariceta B, Agirre X, Urribarri AD, Ayala R, Martínez-Cuadrón D, Bergua JM, Vives S, Algarra L, Tormo M, Martínez P, Serrano J, Simoes C, Herrera P, Calasanz MJ, Alfonso-Piérola A, Paiva B, Martínez-López J, San Miguel JF, Prósper F, Montesinos P. The transcriptomic landscape of elderly acute myeloid leukemia identifies B7H3 and BANP as a favorable signature in high-risk patients. Front Oncol 2022; 12:1054458. [PMID: 36505804 PMCID: PMC9729799 DOI: 10.3389/fonc.2022.1054458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) in the elderly remains a clinical challenge, with a five-year overall survival rate below 10%. The current ELN 2017 genetic risk classification considers cytogenetic and mutational characteristics to stratify fit AML patients into different prognostic groups. However, this classification is not validated for elderly patients treated with a non-intensive approach, and its performance may be suboptimal in this context. Indeed, the transcriptomic landscape of AML in the elderly has been less explored and it might help stratify this group of patients. In the current study, we analyzed the transcriptome of 224 AML patients > 65 years-old at diagnosis treated in the Spanish PETHEMA-FLUGAZA clinical trial in order to identify new prognostic biomarkers in this population. We identified a specific transcriptomic signature for high-risk patients with mutated TP53 or complex karyotype, revealing that low expression of B7H3 gene with high expression of BANP gene identifies a subset of high-risk AML patients surviving more than 12 months. This result was further validated in the BEAT AML cohort. This unique signature highlights the potential of transcriptomics to identify prognostic biomarkers in in elderly AML.
Collapse
Affiliation(s)
- Sara Villar
- Servicio de Hematología y Terapia Celular, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain,CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Beñat Ariceta
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain,Centro de Investigación Médica Aplicada (CIMA) LAB Diagnostics, Universidad de Navarra, Pamplona, Spain,Program of Hematology-Oncology, CIMA, Universidad de Navarra, Pamplona, Spain
| | - Xabier Agirre
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain,Program of Hematology-Oncology, CIMA, Universidad de Navarra, Pamplona, Spain
| | | | - Rosa Ayala
- Hospital Universitario 12 de octubre, Madrid, Spain
| | | | | | - Susana Vives
- ICO Badalona- Hospital Germans Trias i Pujol, Badalona, Spain
| | | | - Mar Tormo
- Hospital Clínico Universitario de Valencia, Valencia, Spain
| | | | - Josefina Serrano
- Hospital Universitario Reina Sofía, Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Catia Simoes
- Program of Hematology-Oncology, CIMA, Universidad de Navarra, Pamplona, Spain
| | | | - Maria José Calasanz
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain,Centro de Investigación Médica Aplicada (CIMA) LAB Diagnostics, Universidad de Navarra, Pamplona, Spain
| | - Ana Alfonso-Piérola
- Servicio de Hematología y Terapia Celular, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain,CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Bruno Paiva
- Servicio de Hematología y Terapia Celular, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain,CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain,Centro de Investigación Médica Aplicada (CIMA) LAB Diagnostics, Universidad de Navarra, Pamplona, Spain
| | | | - Jesús F. San Miguel
- Servicio de Hematología y Terapia Celular, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain,CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain
| | - Felipe Prósper
- Servicio de Hematología y Terapia Celular, Clínica Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IDISNA), Pamplona, Spain,CIBERONC Centro de Investigación Biomédica en Red de Cáncer, Pamplona, Spain,*Correspondence: Felipe Prósper, ; Pau Montesinos,
| | - Pau Montesinos
- Hospital Universitario y Politécnico la Fe, Valencia, Spain,*Correspondence: Felipe Prósper, ; Pau Montesinos,
| |
Collapse
|
10
|
Tyagi A, Ly S, El-Dana F, Yuan B, Jaggupilli A, Grimm S, Konopleva M, Bühring HJ, Battula VL. Evidence supporting a role for the immune checkpoint protein B7-H3 in NK cell-mediated cytotoxicity against AML. Blood 2022; 139:2782-2796. [PMID: 35231101 PMCID: PMC11022957 DOI: 10.1182/blood.2021014671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
We observed that the immune checkpoint protein B7-H3 is overexpressed in acute myeloid leukemia (AML) patients with poor treatment outcomes. Inhibition of B7-H3 expression or blocking of its activity using a novel monoclonal antibody (T-1A5) in AML cells significantly enhanced natural killer (NK) cell-mediated cytotoxicity in AML cells in vitro and in vivo. Moreover, a human-mouse chimera of this antibody (ChT-1A5) induced antibody-dependent cell-mediated cytotoxicity (ADCC) in B7-H3+ primary AML cells, but not in normal hematopoietic cells, suggesting the specify of this antibody for AML cells. Epitope mapping studies identified that both T-1A5 and ChT-1A5 antibodies bind to the FG-loop region of B7-H3, which is known to regulate the immunosuppressive function of B7-H3. Furthermore, treatment with ChT-1A5 in combination with human NK cells significantly prolonged survival in AML patient-derived xenograft (PDX) models. Our results suggest that the ChT-1A5 antibody can inhibit the immunosuppressive function of B7-H3 protein as well as induce ADCC in B7-H3+ AML.
Collapse
Affiliation(s)
- Anudishi Tyagi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stanley Ly
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Fouad El-Dana
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Bin Yuan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Appalaraju Jaggupilli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - V. Lokesh Battula
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
11
|
Ogt Demonstrated Conspicuous Clinical Significance in Cancers, from Pan-Cancer to Small-Cell Lung Cancer. JOURNAL OF ONCOLOGY 2022; 2022:2010341. [PMID: 35356257 PMCID: PMC8959957 DOI: 10.1155/2022/2010341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/18/2022] [Indexed: 11/25/2022]
Abstract
The clinical progression of small-cell lung cancer (SCLC) remains pessimistic. The aim of the present study was to promote the understanding of the clinical significance and mechanism of O-linked N-acetylglucosamine (GlcNAc) transferase (OGT) in SCLC. Wilcoxon tests, standardized mean difference (SMD), and Kruskal–Wallis tests were utilized to compare OGT level differences among the experimental and control groups. The univariate Cox regression analysis, Kaplan–Meier curves, and receiver operating characteristic curves were applied to determine OGT's clinical relevance in cancers. The Spearman correlation analysis and enrichment analysis were utilized to explore the underlying mechanisms of OGT in cancers. For the first time in the field, we provide an overview of OGT in 32 cancers using a large number of samples (n = 21,196), determining distinct OGT expression in 25 cancers and its prognosis effects in 12 cancers. Furthermore, using 950 samples from multiple sources, upregulated OGT was found in both mRNA and protein levels in SCLC (SMD = 0.93, 95% CI [0.24, 1.63]). Higher OGT levels represented a more unfavorable disease-free interval for SCLC patients (p < 0.001). The research also identified OGT expression as a potential marker for SCLC prediction (sensitivity = 0.79, specificity = 0.86, and AUC = 0.88). The high expression of OGT in SCLC may result from the positive regulation of two transcription factors—DEK and XRN2. We primarily investigated the underlying mechanisms of OGT in SCLC. Herein, based on the analyses from pan-cancer to SCLC, OGT demonstrated conspicuous clinical significance. OGT may be an underlying biomarker for the treatment and identification of some cancers, including SCLC.
Collapse
|
12
|
Zhang LY, Jin Y, Xia PH, Lin J, Ma JC, Li T, Liu ZQ, Xiang HL, Cheng C, Xu ZJ, Zhou H, Qian J. Integrated analysis reveals distinct molecular, clinical, and immunological features of B7-H3 in acute myeloid leukemia. Cancer Med 2021; 10:7831-7846. [PMID: 34562306 PMCID: PMC8559480 DOI: 10.1002/cam4.4284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 06/25/2021] [Accepted: 08/29/2021] [Indexed: 12/12/2022] Open
Abstract
The role of B7‐H3 in acute myeloid leukemia (AML) is not fully understood. Two previous studies investigating its expression and significances in AML are partially different. In this study, we aimed to systematically characterize the genomic and immune landscape in AML patients with altered B7‐H3 expression using multi‐omics data in the public domain. We found significantly increased B7‐H3 expression in AML compared to either other hematological malignancies or healthy controls. Clinically, high B7‐H3 expression was associated with old age, TP53 mutations, wild‐type WT1 and CEBPA, and the M3 and M5 FAB subtypes. Moreover, we observed that increased B7‐H3 expression correlated significantly with a poor outcome of AML patients in four independent datasets. Gene set enrichment analysis (GSEA) revealed the enrichment of the “EMT” oncogenic gene signatures in high B7‐H3 expressers. Further investigation suggested that B7‐H3 was more likely to be associated with immune‐suppressive cells (macrophages, neutrophils, dendritic cells, and Th17 cells). B7‐H3 was also positively associated with a number of checkpoint genes, such as VISTA (B7‐H5), CD80 (B7‐1), CD86 (B7‐2), and CD70. In summary, we uncovered distinct genomic and immunologic features associated with B7‐H3 expression in AML. This may lead to a better understanding of the molecular mechanisms underlying B7‐H3 dysregulation in AML and to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Ling-Yi Zhang
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Jiangsu, China.,Zhenjiang Clinical Research Center of Hematology, Jiangsu, China
| | - Ye Jin
- Zhenjiang Clinical Research Center of Hematology, Jiangsu, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Jiangsu, China
| | - Pei-Hui Xia
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Jiangsu, China.,Zhenjiang Clinical Research Center of Hematology, Jiangsu, China
| | - Jiang Lin
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Jiangsu, China.,Zhenjiang Clinical Research Center of Hematology, Jiangsu, China
| | - Ji-Chun Ma
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Jiangsu, China.,Zhenjiang Clinical Research Center of Hematology, Jiangsu, China
| | - Ting Li
- Zhenjiang Clinical Research Center of Hematology, Jiangsu, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Jiangsu, China
| | - Zi-Qi Liu
- Zhenjiang Clinical Research Center of Hematology, Jiangsu, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Jiangsu, China
| | - He-Lin Xiang
- Zhenjiang Clinical Research Center of Hematology, Jiangsu, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Jiangsu, China
| | - Chen Cheng
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Jiangsu, China.,Zhenjiang Clinical Research Center of Hematology, Jiangsu, China
| | - Zi-Jun Xu
- Laboratory Center, Affiliated People's Hospital of Jiangsu University, Jiangsu, China.,Zhenjiang Clinical Research Center of Hematology, Jiangsu, China
| | - Hong Zhou
- School of Medical Science and Laboratory Medicine, Jiangsu University, Jiangsu, China
| | - Jun Qian
- Zhenjiang Clinical Research Center of Hematology, Jiangsu, China.,Department of Hematology, Affiliated People's Hospital of Jiangsu University, Jiangsu, China
| |
Collapse
|