1
|
Wang Y, Yang X, Liu Y, Li Y. A review of common immunotherapy and nano immunotherapy for acute myeloid leukemia. Front Immunol 2025; 16:1505247. [PMID: 40129984 PMCID: PMC11931025 DOI: 10.3389/fimmu.2025.1505247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy. Traditional chemotherapy methods not only bring serious side effects, but also lead to high recurrence rate and drug resistance in some patients. However, as an emerging therapeutic strategy, immunotherapy has shown great potential in the field of AML treatment in recent years. At present, common immunotherapy methods for AML include monoclonal antibodies, CAR-T cell therapy, and immune checkpoint inhibitors. With the deepening of research and technological progress, especially the application of nanotechnology in medicine, new immunotherapy is expected to become one of the important means for the treatment of acute myeloid leukemia in the future.
Collapse
Affiliation(s)
- Yaoyao Wang
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| | - Xiancong Yang
- Laboratory Department, Qilu Hospital of ShanDong University Dezhou Hospital, Dezhou, Shandong, China
| | - Yalin Liu
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
2
|
Lessiak U, Brandstoetter T, Nell B, Klein K, Mlynek G, Wimmer L, Scheiblecker L, Tichy A, Hoelbl-Kovacic A. Preserved bevacizumab (Avastin®) eye drops for application in multidose containers - an in-vitro characterisation. BMC Vet Res 2025; 21:129. [PMID: 40025446 PMCID: PMC11871757 DOI: 10.1186/s12917-025-04592-4] [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: 03/23/2023] [Accepted: 02/11/2025] [Indexed: 03/04/2025] Open
Abstract
PURPOSE Monoclonal antibodies have made an immense contribution to the treatment of various human diseases. We aimed at investigating an affordable treatment option for veterinary patients with corneal neovascularization by adding the preservative benzalkonium chloride (BAC) to bevacizumab (Avastin®) for usage in multidose containers. A comprehensive analytical similarity assessment of preserved and unpreserved bevacizumab after dilution and storage was carried out. METHODS Diluted and preserved bevacizumab was analysed at different time points for a 4-week period and compared with unpreserved bevacizumab at the same concentrations at each time point. Native-PAGE, immunoblotting and HP-SEC were used to observe aggregation and degradation. DLS provided information about particle size and dispersity. Bevacizumab quantified by ELISA was conducted to determine its biological activity. Dose response curves and cell migration assays were performed to detect possible toxic effects and determine biological activity and efficacy of the drug using HUVECs. RESULTS Native-PAGE, immunoblotting and HP-SEC analysis did not show any changes or degradation products in the presence of BAC and after storage compared to unpreserved bevacizumab. The overlapping intensity-based particle size distribution obtained from DLS showed similarity in all tested groups and homogeneity was maintained. ELISA accurately detected bevacizumab at different concentrations. HUVECs incubated with preserved or unpreserved bevacizumab showed a comparable effect on cell migration. No decrease in cell viability was detected. CONCLUSION Equivalence tests demonstrated that bevacizumab is stable after dilution, storage and preservation with BAC. Our study shows that preserved bevacizumab applied in mutidose containers can be considered as a cost-effective alternative to the otherwise single-dose treatments.
Collapse
Affiliation(s)
- Ulrike Lessiak
- Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria.
| | - Tania Brandstoetter
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria
| | - Barbara Nell
- Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria
| | - Klara Klein
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria
| | - Georg Mlynek
- BOKU Core Facility Biomolecular & Cellular Analysis, BOKU - University of Natural Resources and Life Sciences, Muthgasse 11, Vienna, 1190, Austria
| | - Lukas Wimmer
- Division of Pharmaceutical Technology and Biopharmaceutics, University of Vienna, Josef-Holaubek-Platz 2, Vienna, 1090, Austria
| | - Lisa Scheiblecker
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria
| | - Alexander Tichy
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Veterinaerplatz 1, Vienna, 1210, Austria
| |
Collapse
|
3
|
Wang J, Wang Y, Jiang X. Targeting anticancer immunity in melanoma tumour microenvironment: unleashing the potential of adjuvants, drugs, and phytochemicals. J Drug Target 2024; 32:1052-1072. [PMID: 39041142 DOI: 10.1080/1061186x.2024.2384071] [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: 05/08/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Melanoma poses a challenge in oncology because of its aggressive nature and limited treatment modalities. The tumour microenvironment (TME) in melanoma contains unique properties such as an immunosuppressive and high-density environment, unusual vasculature, and a high number of stromal and immunosuppressive cells. In recent years, numerous experiments have focused on boosting the immune system to effectively remove malignant cells. Adjuvants, consisting of phytochemicals, toll-like receptor (TLR) agonists, and cytokines, have shown encouraging results in triggering antitumor immunity and augmenting the therapeutic effectiveness of anticancer therapy. These adjuvants can stimulate the maturation of dendritic cells (DCs) and infiltration of cytotoxic CD8+ T lymphocytes (CTLs). Furthermore, nanocarriers can help to deliver immunomodulators and antigens directly to the tumour stroma, thereby improving their efficacy against malignant cells. The remodelling of melanoma TME utilising phytochemicals, agonists, and other adjuvants can be combined with current modalities for improving therapy outcomes. This review article explores the potential of adjuvants, drugs, and their nanoformulations in enhancing the anticancer potency of macrophages, CTLs, and natural killer (NK) cells. Additionally, the capacity of these agents to repress the function of immunosuppressive components of melanoma TME, such as immunosuppressive subsets of macrophages, stromal and myeloid cells will be discussed.
Collapse
Affiliation(s)
- Jingping Wang
- Emergency Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Yaping Wang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| | - Xiaofang Jiang
- Respiratory and Oncology Department, Zhejiang Provincial General Hospital of the Chinese People's Armed Police Force, Zhejiang, China
| |
Collapse
|
4
|
Wang Z, Liu M, Yang Q. Glutamine and leukemia research: progress and clinical prospects. Discov Oncol 2024; 15:391. [PMID: 39215845 PMCID: PMC11365919 DOI: 10.1007/s12672-024-01245-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Leukemia is an abnormal proliferation of white blood cells that occurs in bone marrow and expands through the blood. It arises from dysregulated differentiation, uncontrolled growth, and inhibition of apoptosis. Glutamine (GLN) is a "conditionally essential" amino acid that promotes growth and proliferation of leukemic cells. Recently, details about the role of GLN and its metabolism in the diagnosis and treatment of acute myeloid, chronic lymphocytic, and acute lymphoblastic leukemia have emerged. The uptake of GLN by leukemia cells and the dynamic changes of glutamine-related indexes in leukemia patients may be able to assist in determining whether the condition of leukemia is in a state of progression, remission or relapse. Utilizing the possible differences in GLN metabolism in different subtypes of leukemia may help to differentiate between different subtypes of leukemia, thus providing a basis for accurate diagnosis. Targeting GLN metabolism in leukemia requires simultaneous blockade of multiple metabolic pathways without interfering with the normal cellular and immune functions of the body to achieve effective leukemia therapy. The present review summarizes recent advances, possible applications, and clinical perspectives of GLN metabolism in leukemia. In particular, it focuses on the prospects of GLN metabolism in the diagnosis and treatment of acute myeloid leukemia. The review provides new directions and hints at potential roles for future clinical treatments and studies.
Collapse
Affiliation(s)
- Zexin Wang
- Mianyang Central Hospital, Fucheng District, Mianyang, 621000, Sichuan, China.
| | - Miao Liu
- Mianyang Central Hospital, Fucheng District, Mianyang, 621000, Sichuan, China
| | - Qiang Yang
- Mianyang Central Hospital, Fucheng District, Mianyang, 621000, Sichuan, China
| |
Collapse
|
5
|
Bianchi M, Reichen C, Croset A, Fischer S, Eggenschwiler A, Grübler Y, Marpakwar R, Looser T, Spitzli P, Herzog C, Villemagne D, Schiegg D, Abduli L, Iss C, Neculcea A, Franchini M, Lekishvili T, Ragusa S, Zitt C, Kaufmann Y, Auge A, Hänggi M, Ali W, Frasconi TM, Wullschleger S, Schlegel I, Matzner M, Lüthi U, Schlereth B, Dawson KM, Kirkin V, Ochsenbein AF, Grimm S, Reschke N, Riether C, Steiner D, Leupin N, Goubier A. The CD33xCD123xCD70 Multispecific CD3-Engaging DARPin MP0533 Induces Selective T Cell-Mediated Killing of AML Leukemic Stem Cells. Cancer Immunol Res 2024; 12:921-943. [PMID: 38683145 PMCID: PMC11217734 DOI: 10.1158/2326-6066.cir-23-0692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/04/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
The prognosis of patients with acute myeloid leukemia (AML) is limited, especially for elderly or unfit patients not eligible for hematopoietic stem cell (HSC) transplantation. The disease is driven by leukemic stem cells (LSCs), which are characterized by clonal heterogeneity and resistance to conventional therapy. These cells are therefore believed to be a major cause of progression and relapse. We designed MP0533, a multispecific CD3-engaging designed ankyrin repeat protein (DARPin) that can simultaneously bind to three antigens on AML cells (CD33, CD123, and CD70), aiming to enable avidity-driven T cell-mediated killing of AML cells coexpressing at least two of the antigens. In vitro, MP0533 induced selective T cell-mediated killing of AML cell lines, as well as patient-derived AML blasts and LSCs, expressing two or more target antigens, while sparing healthy HSCs, blood, and endothelial cells. The higher selectivity also resulted in markedly lower levels of cytokine release in normal human blood compared to single antigen-targeting T-cell engagers. In xenograft AML mice models, MP0533 induced tumor-localized T-cell activation and cytokine release, leading to complete eradication of the tumors while having no systemic adverse effects. These studies show that the multispecific-targeting strategy used with MP0533 holds promise for improved selectivity toward LSCs and efficacy against clonal heterogeneity, potentially bringing a new therapeutic option to this group of patients with a high unmet need. MP0533 is currently being evaluated in a dose-escalation phase 1 study in patients with relapsed or refractory AML (NCT05673057).
Collapse
Affiliation(s)
| | | | - Amelie Croset
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | | | | | - Thamar Looser
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | | | | | | | - Chloé Iss
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | | | - Simone Ragusa
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Christof Zitt
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | - Alienor Auge
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Martin Hänggi
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Waleed Ali
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | | | - Iris Schlegel
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | | | - Ursina Lüthi
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | | | | | - Adrian F. Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | - Nina Reschke
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | | | | | - Anne Goubier
- Molecular Partners AG, Zurich-Schlieren, Switzerland.
| |
Collapse
|
6
|
Bordeleau ME, Audemard É, Métois A, Theret L, Lisi V, Farah A, Spinella JF, Chagraoui J, Moujaber O, Aubert L, Khakipoor B, Mallinger L, Boivin I, Mayotte N, Hajmirza A, Bonneil É, Béliveau F, Pfammatter S, Feghaly A, Boucher G, Gendron P, Thibault P, Barabé F, Lemieux S, Richard-Carpentier G, Hébert J, Lavallée VP, Roux PP, Sauvageau G. Immunotherapeutic targeting of surfaceome heterogeneity in AML. Cell Rep 2024; 43:114260. [PMID: 38838225 DOI: 10.1016/j.celrep.2024.114260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/29/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
Immunotherapy remains underexploited in acute myeloid leukemia (AML) compared to other hematological malignancies. Currently, gemtuzumab ozogamicin is the only therapeutic antibody approved for this disease. Here, to identify potential targets for immunotherapeutic intervention, we analyze the surface proteome of 100 genetically diverse primary human AML specimens for the identification of cell surface proteins and conduct single-cell transcriptome analyses on a subset of these specimens to assess antigen expression at the sub-population level. Through this comprehensive effort, we successfully identify numerous antigens and markers preferentially expressed by primitive AML cells. Many identified antigens are targeted by therapeutic antibodies currently under clinical evaluation for various cancer types, highlighting the potential therapeutic value of the approach. Importantly, this initiative uncovers AML heterogeneity at the surfaceome level, identifies several antigens and potential primitive cell markers characterizing AML subgroups, and positions immunotherapy as a promising approach to target AML subgroup specificities.
Collapse
Affiliation(s)
- Marie-Eve Bordeleau
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| | - Éric Audemard
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Arnaud Métois
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Louis Theret
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Véronique Lisi
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Azer Farah
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Jean-François Spinella
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Jalila Chagraoui
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Ossama Moujaber
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Léo Aubert
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Banafsheh Khakipoor
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada
| | - Laure Mallinger
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Isabel Boivin
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Nadine Mayotte
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Azadeh Hajmirza
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Éric Bonneil
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - François Béliveau
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada
| | - Sybille Pfammatter
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Albert Feghaly
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Geneviève Boucher
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Patrick Gendron
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Pierre Thibault
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Chemistry, Faculty of Arts and Science, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Frédéric Barabé
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC G1V 4G2, Canada
| | - Sébastien Lemieux
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| | - Guillaume Richard-Carpentier
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; Department of Medicine, Division of Medical Oncology and Hematology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Josée Hébert
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada.
| | - Vincent-Philippe Lavallée
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Centre Hospitalier Universitaire Sainte-Justine Research Center, Montréal, QC H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Hematology and Oncology Division, Centre Hospitalier Universitaire Sainte-Justine, Montréal, QC H3T 1C5, Canada.
| | - Philippe P Roux
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| | - Guy Sauvageau
- The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC H3T 1J4, Canada; Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Division of Hematology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, QC H1T 2M4, Canada.
| |
Collapse
|
7
|
Gonçalves AC, Alves R, Sarmento-Ribeiro AB. Advancements in Biomarkers and Molecular Targets in Hematological Neoplasias. Int J Mol Sci 2024; 25:6570. [PMID: 38928276 PMCID: PMC11203996 DOI: 10.3390/ijms25126570] [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: 05/25/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Hematological neoplasias are among the most common cancers worldwide, and the number of new cases has been on the rise since 1990, reaching 1 [...].
Collapse
Affiliation(s)
- Ana Cristina Gonçalves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Raquel Alves
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
| | - Ana Bela Sarmento-Ribeiro
- Laboratory of Oncobiology and Hematology (LOH), University Clinics of Hematology and Oncology, Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal; (R.A.); (A.B.S.-R.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR)—Group of Environmental Genetics of Oncobiology (CIMAGO), FMUC, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Hematology Service, Centro Hospitalar Universitário de Coimbra, Unidade Local de Saúde de Coimbra, 3000-061 Coimbra, Portugal
| |
Collapse
|
8
|
Pereira MP, Herrity E, Kim DDH. TP53-mutated acute myeloid leukemia and myelodysplastic syndrome: biology, treatment challenges, and upcoming approaches. Ann Hematol 2024; 103:1049-1067. [PMID: 37770618 DOI: 10.1007/s00277-023-05462-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
Improved understanding of TP53 biology and the clinicopathological features of TP53-mutated myeloid neoplasms has led to the recognition of TP53-mutated acute myeloid leukemia/myelodysplastic syndrome (TP53m AML/MDS) as a unique entity, characterized by dismal outcomes following conventional therapies. Several clinical trials have investigated combinations of emerging therapies for these patients with the poorest molecular prognosis among myeloid neoplasms. Although some emerging therapies have shown improvement in overall response rates, this has not translated into better overall survival, hence the notion that p53 remains an elusive target. New therapeutic strategies, including novel targeted therapies, immune checkpoint inhibitors, and monoclonal antibodies, represent a shift away from cytotoxic and hypomethylating-based therapies, towards approaches combining non-immune and novel immune therapeutic strategies. The triple combination of azacitidine and venetoclax with either magrolimab or eprenetapopt have demonstrated safety in early trials, with phase III trials currently underway, and promising interim clinical results. This review compiles background on TP53 biology, available and emerging therapies along with their mechanisms of action for the TP53m disease entity, current treatment challenges, and recently published data and status of ongoing clinical trials for TP53m AML/MDS.
Collapse
Affiliation(s)
- Mariana Pinto Pereira
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Elizabeth Herrity
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Dennis D H Kim
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada.
- Leukemia Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Hematology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Zhu Q, Yang Y, Chen K, Zhang Q, Huang Y, Jian S. Diffuse large B-cell lymphoma: the significance of CD8 + tumor-infiltrating lymphocytes exhaustion mediated by TIM3/Galectin-9 pathway. J Transl Med 2024; 22:174. [PMID: 38369502 PMCID: PMC10874540 DOI: 10.1186/s12967-024-05002-3] [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: 01/15/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Overexpression of T-cell immunoglobulin and mucin domain-containing protein 3 (TIM3) is related to the exhaustion of CD8+ tumor-infiltrating lymphocytes (TILs) in diffuse large B-cell lymphoma (DLBCL). However, the mechanism of TIM3-mediated CD8+TILs exhaustion in DLBCL remains poorly understood. Therefore, we aimed to clarify the potential pathway involved in TIM3-mediated CD8+TILs exhaustion and its significance in DLBCL. METHODS The expression of TIM3 and its correlation with CD8+TILs exhaustion, the key ligand of TIM3, and the potential pathway of TIM3-mediated CD8+TILs exhaustion in DLBCL were analyzed using single-cell RNA sequencing and validated by RNA sequencing. The biological significance of TIM3-related pathway in DLBCL was investigated based on RNA sequencing, immunohistochemistry, and reverse transcription-quantitative polymerase chain reaction data. Finally, the possible regulatory mechanism of TIM3-related pathway in DLBCL was explored using single-cell RNA sequencing and RNA sequencing. RESULTS Our results demonstrated that CD8+TILs, especially the terminally exhausted state, were the major clusters that expressed TIM3 in DLBCL. Galectin-9, mainly expressed in M2 macrophages, is the key ligand of TIM3 and can induce the exhaustion of CD8+TILs through TIM3/Galectin-9 pathway. Meanwhile, high TIM3/Galectin-9 enrichment is related to immunosuppressive tumor microenvironment, severe clinical manifestations, inferior prognosis, and poor response to CHOP-based chemotherapy, and can predict the clinical efficacy of immune checkpoint blockade therapy in DLBCL. Furthermore, the TIM3/Galectin-9 enrichment in DLBCL may be regulated by the IFN-γ signaling pathway. CONCLUSIONS Our study highlights that TIM3/Galectin-9 pathway plays a crucial role in CD8+TILs exhaustion and the immune escape of DLBCL, which facilitates further functional studies and could provide a theoretical basis for the development of novel immunotherapy in DLBCL.
Collapse
Affiliation(s)
- Qiqi Zhu
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan Nan Road, Nanchong, 637000, Sichuan, China
| | - Yiming Yang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan Nan Road, Nanchong, 637000, Sichuan, China
| | - Kexin Chen
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan Nan Road, Nanchong, 637000, Sichuan, China
| | - Qiaoyu Zhang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan Nan Road, Nanchong, 637000, Sichuan, China
| | - Yifan Huang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan Nan Road, Nanchong, 637000, Sichuan, China
| | - Shunhai Jian
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, No. 1 Maoyuan Nan Road, Nanchong, 637000, Sichuan, China.
| |
Collapse
|
10
|
Zhang M, Liu C, Li Y, Li H, Zhang W, Liu J, Wang L, Sun C. Galectin-9 in cancer therapy: from immune checkpoint ligand to promising therapeutic target. Front Cell Dev Biol 2024; 11:1332205. [PMID: 38264357 PMCID: PMC10803597 DOI: 10.3389/fcell.2023.1332205] [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: 11/02/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Galectin-9 (Gal-9) is a vital member of the galectin family, functioning as a multi-subtype galactose lectin with diverse biological roles. Recent research has revealed that Gal-9's interaction with tumors is an independent factor that influences tumor progression. Furthermore, Gal-9 in the immune microenvironment cross-talks with tumor-associated immune cells, informing the clarification of Gal-9's identity as an immune checkpoint. A thorough investigation into Gal-9's role in various cancer types and its interaction with the immune microenvironment could yield novel strategies for subsequent targeted immunotherapy. This review focuses on the latest advances in understanding the direct and indirect cross-talk between Gal-9 and hematologic malignancies, in addition to solid tumors. In addition, we discuss the prospects of Gal-9 in tumor immunotherapy, including its cross-talk with the ligand TIM-3 and its potential in immune-combination therapy.
Collapse
Affiliation(s)
- Minpu Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Wenfeng Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jingyang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Liquan Wang
- Department of Thyroid and Breast Surgery, Weifang People’s Hospital, Weifang, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
11
|
Gera K, Chauhan A, Castillo P, Rahman M, Mathavan A, Mathavan A, Oganda-Rivas E, Elliott L, Wingard JR, Sayour EJ. Vaccines: a promising therapy for myelodysplastic syndrome. J Hematol Oncol 2024; 17:4. [PMID: 38191498 PMCID: PMC10773074 DOI: 10.1186/s13045-023-01523-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/23/2023] [Indexed: 01/10/2024] Open
Abstract
Myelodysplastic neoplasms (MDS) define clonal hematopoietic malignancies characterized by heterogeneous mutational and clinical spectra typically seen in the elderly. Curative treatment entails allogeneic hematopoietic stem cell transplant, which is often not a feasible option due to older age and significant comorbidities. Immunotherapy has the cytotoxic capacity to elicit tumor-specific killing with long-term immunological memory. While a number of platforms have emerged, therapeutic vaccination presents as an appealing strategy for MDS given its promising safety profile and amenability for commercialization. Several preclinical and clinical trials have investigated the efficacy of vaccines in MDS; these include peptide vaccines targeting tumor antigens, whole cell-based vaccines and dendritic cell-based vaccines. These therapeutic vaccines have shown acceptable safety profiles, but consistent clinical responses remain elusive despite robust immunological reactions. Combining vaccines with immunotherapeutic agents holds promise and requires further investigation. Herein, we highlight therapeutic vaccine trials while reviewing challenges and future directions of successful vaccination strategies in MDS.
Collapse
Affiliation(s)
- Kriti Gera
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Anjali Chauhan
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Immunotherapy, University of Florida, Gainesville, FL, USA
| | - Paul Castillo
- Division of Hematology and Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Maryam Rahman
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Immunotherapy, University of Florida, Gainesville, FL, USA
| | - Akash Mathavan
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Akshay Mathavan
- Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Elizabeth Oganda-Rivas
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Immunotherapy, University of Florida, Gainesville, FL, USA
| | - Leighton Elliott
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - John R Wingard
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA.
| | - Elias J Sayour
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Immunotherapy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Knorr K, Rahman J, Erickson C, Wang E, Monetti M, Li Z, Ortiz-Pacheco J, Jones A, Lu SX, Stanley RF, Baez M, Fox N, Castro C, Marino AE, Jiang C, Penson A, Hogg SJ, Mi X, Nakajima H, Kunimoto H, Nishimura K, Inoue D, Greenbaum B, Knorr D, Ravetch J, Abdel-Wahab O. Systematic evaluation of AML-associated antigens identifies anti-U5 SNRNP200 therapeutic antibodies for the treatment of acute myeloid leukemia. NATURE CANCER 2023; 4:1675-1692. [PMID: 37872381 PMCID: PMC10733148 DOI: 10.1038/s43018-023-00656-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 09/19/2023] [Indexed: 10/25/2023]
Abstract
Despite recent advances in the treatment of acute myeloid leukemia (AML), there has been limited success in targeting surface antigens in AML, in part due to shared expression across malignant and normal cells. Here, high-density immunophenotyping of AML coupled with proteogenomics identified unique expression of a variety of antigens, including the RNA helicase U5 snRNP200, on the surface of AML cells but not on normal hematopoietic precursors and skewed Fc receptor distribution in the AML immune microenvironment. Cell membrane localization of U5 snRNP200 was linked to surface expression of the Fcγ receptor IIIA (FcγIIIA, also known as CD32A) and correlated with expression of interferon-regulated immune response genes. Anti-U5 snRNP200 antibodies engaging activating Fcγ receptors were efficacious across immunocompetent AML models and were augmented by combination with azacitidine. These data provide a roadmap of AML-associated antigens with Fc receptor distribution in AML and highlight the potential for targeting the AML cell surface using Fc-optimized therapeutics.
Collapse
Affiliation(s)
- Katherine Knorr
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA
| | - Jahan Rahman
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caroline Erickson
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric Wang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Mara Monetti
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoning Li
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliana Ortiz-Pacheco
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Jones
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA
| | - Sydney X Lu
- Stanford University School of Medicine, Stanford, CA, USA
| | - Robert F Stanley
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria Baez
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA
| | - Nina Fox
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cynthia Castro
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alessandra E Marino
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA
| | - Caroline Jiang
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA
| | - Alex Penson
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon J Hogg
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaoli Mi
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hiroyoshi Kunimoto
- Department of Stem Cell and Immune Regulation, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Benjamin Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David Knorr
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA
| | - Jeffrey Ravetch
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY, USA.
| | - Omar Abdel-Wahab
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
13
|
Stengel A, Meggendorfer M, Walter W, Baer C, Nadarajah N, Hutter S, Kern W, Haferlach T, Haferlach C. Interplay of TP53 allelic state, blast count, and complex karyotype on survival of patients with AML and MDS. Blood Adv 2023; 7:5540-5548. [PMID: 37505914 PMCID: PMC10515307 DOI: 10.1182/bloodadvances.2023010312] [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: 03/27/2023] [Revised: 07/06/2023] [Accepted: 07/26/2023] [Indexed: 07/30/2023] Open
Abstract
Several clinical and genetic factors impact overall survival (OS) in myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML), including complex karyotype (CK), TP53 allelic state, and blast count. We analyzed the interplay of these factors by performing Cox regression analysis and by determining the frequency of TP53 single-hit (sh) and double-hit (dh) events and OS in MDS (n = 747) with <5% blasts, with ≥5% but <10% blasts, and ≥10% but <20% blasts and AML (n = 772). MDS with <5% blasts showed the best outcome, followed by with ≥5% but <10% blasts, and ≥10% but <20% blasts, and AML (median OS: 75, 54, 27, and 18 months, respectively). The same hierarchy was observed when each subgroup was divided into TP53sh, TP53dh, and without TP53 alterations (alt), revealing a dismal outcome of TP53dh in all subgroups (17, 10, 8, and 1 month[s], respectively). MDS with <5% blasts differed from the other subgroups by showing predominantly TP53sh (76% of TP53alt cases), and by an independent adverse impact of CK on OS (hazard ratio, 5.2; P < .001). The remaining subgroups displayed many similarities, with TP53dh found at high frequencies (67%, 91%, and 71%, respectively) and only TP53alt but not CK independently influencing OS, and TP53dh showing the strongest influence. When the total cohort was split based on TP53 state, only the blast count and not CK had an independent adverse impact on OS in all subgroups. Thus, TP53dh is the strongest prognostic factor, further supporting its integration into risk stratification guidelines and classification as a separate entity. However, the blast count also influences OS independent of TP53 state, whereas CK plays a minor prognostic role.
Collapse
|
14
|
Braun T, Schrader A. Education and Empowering Special Forces to Eradicate Secret Defectors: Immune System-Based Treatment Approaches for Mature T- and NK-Cell Malignancies. Cancers (Basel) 2023; 15:cancers15092532. [PMID: 37173999 PMCID: PMC10177197 DOI: 10.3390/cancers15092532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Mature T- and NK-cell leukemia/lymphoma (MTCL/L) constitute a heterogeneous group of, currently, 30 distinct neoplastic entities that are overall rare, and all present with a challenging molecular markup. Thus, so far, the use of first-line cancer treatment modalities, including chemotherapies, achieve only limited clinical responses associated with discouraging prognoses. Recently, cancer immunotherapy has evolved rapidly, allowing us to help patients with, e.g., solid tumors and also relapsed/refractory B-cell malignancies to achieve durable clinical responses. In this review, we systematically unveiled the distinct immunotherapeutic approaches available, emphasizing the special impediments faced when trying to employ immune system defense mechanisms to target 'one of their own-gone mad'. We summarized the preclinical and clinical efforts made to employ the various platforms of cancer immunotherapies including antibody-drug conjugates, monoclonal as well as bispecific antibodies, immune-checkpoint blockades, and CAR T cell therapies. We emphasized the challenges to, but also the goals of, what needs to be done to achieve similar successes as seen for B-cell entities.
Collapse
Affiliation(s)
- Till Braun
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases, Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
| | - Alexandra Schrader
- Department I of Internal Medicine, Center for Integrated Oncology, Aachen-Bonn-Cologne-Duesseldorf, Excellence Cluster for Cellular Stress Response and Aging-Associated Diseases, Center for Molecular Medicine Cologne, University of Cologne, 50937 Cologne, Germany
- Lymphoma Immuno Biology Team, Equipe Labellisée LIGUE 2023, Centre International de Recherche en Infectiologie, INSERM U1111-CNRS UMR5308, Faculté de Médecine Lyon-Sud, Hospices Civils de Lyon, Université Claude Bernard Lyon I-ENS de Lyon, 69921 Lyon, France
| |
Collapse
|
15
|
Salvatorelli E, Minotti G, Menna P. New Targeted Drugs for Acute Myeloid Leukemia and Antifungals: Pharmacokinetic Challenges and Opportunities. Chemotherapy 2023; 68:170-182. [PMID: 37004510 DOI: 10.1159/000530447] [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: 01/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a life-threatening disease whose treatment is made difficult by a number of mutations or receptor overexpression in the proliferating cellular clones. Life expectancy of patients diagnosed with new, relapsed-refractory, or secondary AML has been improved by drugs targeted at such moieties. Regrettably, however, clinical use of new AML drugs is complicated by pharmacokinetic interactions with other drugs the patient is exposed to. SUMMARY The most relevant drug-drug interactions (DDI) with clinical implications build on competition for or induction/inhibition of CYP3A4, which is a versatile metabolizer of a plethora of pharmacological agents. Here, we review DDI between AML drugs and the agents used to prevent or treat invasive fungal infections (IFI). The pathophysiology of AML, characterized by functionally defective white blood cells and neutropenic/immunosuppressive effects of concomitant induction chemotherapy, can in fact increase the risk of infectious complications, with IFI causing high rates of morbidity and mortality. Triazole antifungals, such as posaconazole, are strong inhibitors of CYP3A4 and may thus cause patient's overexposure to AML drugs that are metabolized by CYP3A4. We describe potential strategies to minimize the consequences of DDI between triazole antifungals and targeted therapies for AML and the role that collaboration between clinical pharmacologists, hematologists, and clinical or laboratory microbiologists may have in these settings. KEY MESSAGES Therapeutic drug monitoring and clinical pharmacology stewardship could represent two strategies that best express multidisciplinary collaboration for improving patient management.
Collapse
Affiliation(s)
| | - Giorgio Minotti
- Department of Medicine, University Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Pharmacology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Pierantonio Menna
- Research Unit of Clinical Pharmacology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
16
|
Bailly C. Contribution of the TIM-3/Gal-9 immune checkpoint to tropical parasitic diseases. Acta Trop 2023; 238:106792. [PMID: 36509129 DOI: 10.1016/j.actatropica.2022.106792] [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] [Received: 10/21/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Neglected tropical parasitic diseases (NTD) are prevalent in many countries and cost-effective treatments remain urgently needed. Novel approaches have been proposed to address these diseases through an action on immune co-inhibitory checkpoints which are exploited by parasites to evade the immune system. Among these checkpoints, TIM-3 has been shown to play a key role in antiparasitic immunity via a repression and functional attenuation of CD4+ and/or CD8+ T-cells. The present review discusses the role of the TIM-3/galectin-9 checkpoint in seven major NTD: Chagas disease, leishmaniasis and malaria (3 trypanosomatid infections), schistosomiasis, toxoplasmosis, echinococcosis and filariasis (4 helminth infections). In each case, the role of the checkpoint has been analyzed and the use of anti-TIM-3 antibodies evaluated as a potential therapeutic approach. In general, the parasitic infection is coupled with an upregulation of TIM-3 expressed on T cells, but not necessarily with an exhaustion of those T cells. In several cases, the use of anti-TIM-3 antibodies represent a possible strategy to reinforce the clearance and to reduce the parasite load. Promising data have been reported in cases of leishmaniasis, malaria and schistosomiasis, whereas a similar approach proved much less efficient (if not deleterious) in cases of echinococcosis and the Chagas disease. Nevertheless, the TIM-3 checkpoint warrants further consideration as a potential immune target to combat these pathologies, using antibodies or drugs capable of reducing directly or indirectly the expression and function of the checkpoint, to restore an immune control.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, Lille (Wasquehal), 59290, France; University of Lille, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, 3 rue du Professeur Laguesse, BP-83, F-59006, Lille, France.
| |
Collapse
|
17
|
Zhou H, Wang F, Niu T. Prediction of prognosis and immunotherapy response of amino acid metabolism genes in acute myeloid leukemia. Front Nutr 2022; 9:1056648. [PMID: 36618700 PMCID: PMC9815546 DOI: 10.3389/fnut.2022.1056648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background Amino acid (AA) metabolism plays a crucial role in cancer. However, its role in acute myeloid leukemia (AML) is still unavailable. We screened out AA metabolic genes, which related to prognosis, and analyzed their correlation with tumor immune microenvironment in AML. Methods We evaluated 472 amino acid metabolism-related genes in 132 AML patients. The predictive risk model was developed according to differentially expressed genes, univariate Cox and LASSO analyses. We validated the risk signature by survival analysis and independence tests. Single-sample gene set enrichment analysis (ssGSEA), tumor immune microenvironment (TME), tumor mutation burden (TMB), functional enrichment, and the IC50 of drugs were assessed to explore the correlations among the risk model, immunity, and drug sensitivity of AML. Results Six amino acid metabolism-related genes were confirmed to develop the risk model, including TRH, HNMT, TFEB, SDSL, SLC43A2, and SFXN3. The high-risk subgroup had an immune "hot" phenotype and was related to a poor prognosis. The high-risk group was also associated with more activity of immune cells, such as Tregs, had higher expression of some immune checkpoints, including PD1 and CTLA4, and might be more susceptible to immunotherapy. Xenobiotic metabolism, the reactive oxygen species (ROS) pathway, fatty acid metabolism, JAK/STAT3, and the inflammatory response were active in the high-risk subgroup. Furthermore, the high-risk subgroup was sensitive to sorafenib, selumetinib, and entospletinib. ssGSEA discovered that the processes of glutamine, arginine, tryptophan, cysteine, histidine, L-serine, isoleucine, threonine, tyrosine, and L-phenylalanine metabolism were more active in the high-risk subgroup. Conclusion This study revealed that AA metabolism-related genes were correlated with the immune microenvironment of AML patients and could predict the prognosis and immunotherapy response of AML patients.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengjuan Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Niu
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Ting Niu,
| |
Collapse
|
18
|
A Bioinformatics View on Acute Myeloid Leukemia Surface Molecules by Combined Bayesian and ABC Analysis. Bioengineering (Basel) 2022; 9:bioengineering9110642. [DOI: 10.3390/bioengineering9110642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
“Big omics data” provoke the challenge of extracting meaningful information with clinical benefit. Here, we propose a two-step approach, an initial unsupervised inspection of the structure of the high dimensional data followed by supervised analysis of gene expression levels, to reconstruct the surface patterns on different subtypes of acute myeloid leukemia (AML). First, Bayesian methodology was used, focusing on surface molecules encoded by cluster of differentiation (CD) genes to assess whether AML is a homogeneous group or segregates into clusters. Gene expressions of 390 patient samples measured using microarray technology and 150 samples measured via RNA-Seq were compared. Beyond acute promyelocytic leukemia (APL), a well-known AML subentity, the remaining AML samples were separated into two distinct subgroups. Next, we investigated which CD molecules would best distinguish each AML subgroup against APL, and validated discriminative molecules of both datasets by searching the scientific literature. Surprisingly, a comparison of both omics analyses revealed that CD339 was the only overlapping gene differentially regulated in APL and other AML subtypes. In summary, our two-step approach for gene expression analysis revealed two previously unknown subgroup distinctions in AML based on surface molecule expression, which may guide the differentiation of subentities in a given clinical–diagnostic context.
Collapse
|
19
|
Bhatt A, Powell K, Prasad V. The AGILE trial of ivosidenib plus azacitidine versus azacitidine alone: How many limitations is too many? Transl Oncol 2022; 25:101523. [PMID: 36063621 PMCID: PMC9460510 DOI: 10.1016/j.tranon.2022.101523] [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: 06/22/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Post-protocol therapy was suboptimal. The control arm of AGILE does not reflect the current standard of care. The primary endpoint of AGILE was changed, and the sample size was decreased. The AGILE investigators halted the trial early based on a non-primary endpoint.
The AGILE trial compared ivosidenib and azacitidine versus azacitidine for IDH1-mutant acute myeloid leukemia (AML) in elderly patients who were ineligible to receive intensive chemotherapy. While the results of this trial appear encouraging, various concerns become evident from the study design and methodology. First, the AGILE trial did not use post-protocol therapy that met the current standard of care. Second, researchers continued patient enrollment despite knowledge of the survival benefit of azacitidine plus venetoclax shown in the VIALE-A trial, resulting in an inferior control arm. Third, the primary endpoint of AGILE was changed from overall survival (OS) to event-free survival (EFS), and the sample size was reduced to expedite the results. Finally, the trial was halted early based on a non-primary endpoint, which likely led to exaggerated effect size or misleading results. We discuss these limitations and continue to advocate for careful analysis of study design to ensure that appropriate and accurate outcomes are implemented in future studies.
Collapse
Affiliation(s)
- Anjali Bhatt
- College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Kerrington Powell
- College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, United States
| | - Vinay Prasad
- Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, United States.
| |
Collapse
|
20
|
Çınar OE, Erdoğdu B, Karadeniz M, Ünal S, Malkan ÜY, Göker H, Haznedaroğlu İC. Comment on Zamfir et al. Hematologic Malignancies Diagnosed in the Context of the mRNA COVID-19 Vaccination Campaign: A Report of Two Cases. Medicina 2022, 58, 874. Medicina (B Aires) 2022; 58:medicina58111575. [PMID: 36363532 PMCID: PMC9696869 DOI: 10.3390/medicina58111575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/23/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The SARS-CoV-2 spike protein mRNA-based vaccines have prevented countless mortality and morbidity, and have an excellent risk/benefit ratio. However, various adverse events may rarely occur after the BNT162b2 vaccine, like any other medical intervention. The COVID-19 itself and the spike protein produced endogenously by mRNA vaccines may have immunological, microenvironmental, prothrombotic, and neoplastic effects. As a contribution to the published report, we would like to share our experience regarding four cases in which myeloid neoplasms emerged following the vaccination. Conclusions: There is no doubt that vaccination could continue along the lines of established universal recommendations. Meanwhile, all hematological adverse events must be closely monitored and reported. Further efforts should be focused on the probable pathobiological mechanisms and causalities of spike protein-related toxicity and clonal myeloid disorders.
Collapse
|
21
|
Kassouri C, Rodriguez Torres S, Gonzalez Suarez N, Duhamel S, Annabi B. EGCG Prevents the Transcriptional Reprogramming of an Inflammatory and Immune-Suppressive Molecular Signature in Macrophage-like Differentiated Human HL60 Promyelocytic Leukemia Cells. Cancers (Basel) 2022; 14:5065. [PMID: 36291849 PMCID: PMC9599716 DOI: 10.3390/cancers14205065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/07/2022] [Accepted: 10/13/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The promyelocytic leukemia cell differentiation process enables recapitulation of the polarized M1 or M2 macrophage-like phenotype with inflammatory and immune-suppressive properties. While evidence supports the anti-inflammatory effect of dietary-derived epigallocatechin-3-gallate (EGCG), its impact on the onset of immune phenotype molecular signature remains unclear. METHODS Human HL60 promyelocytic cells grown in suspension were differentiated into CD11bHigh/CD14Low adherent macrophages with phorbol 12-myristate 13-acetate (PMA). Gelatin zymography was used to assess the levels of matrix metalloproteinase (MMP)-9, and total RNA was isolated for RNAseq and RT-qPCR assessment of differentially expressed gene levels involved in inflammation and immunity. Protein lysates were used to assess the phosphorylation status of signaling intermediates involved in macrophage-like cell differentiation. RESULTS Cell adhesion and induction of MMP-9 were indicative of HL60 cell differentiation into a macrophage-like phenotype. The extracellular signal-regulated kinase (ERK), glycogen synthase kinase (GSK)-3, p90 ribosomal S6 kinases (RSK), and cAMP-response-element-binding protein (CREB) were all phosphorylated, and EGCG reduced such phosphorylation status. Increases in inflammation and immunity genes included, among others, CCL22, CSF1, CSF2, IL1B, and TNF, which inductions were prevented by EGCG. This was corroborated by unbiased transcriptomic analysis which further highlighted the capacity of EGCG to downregulate the hematopoietic stem cell regulator CBFA2T3. CONCLUSION EGCG inhibits inflammatory signaling crosstalk and prevents the onset of an immune phenotype in macrophage-like differentiated cells.
Collapse
Affiliation(s)
- Celia Kassouri
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| | - Sahily Rodriguez Torres
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| | - Narjara Gonzalez Suarez
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| | - Stéphanie Duhamel
- Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Borhane Annabi
- Laboratoire d’Oncologie Moléculaire, Département de Chimie, and CERMO-FC, Université du Québec à Montréal, Montreal, QC H2X 2J6, Canada
| |
Collapse
|
22
|
Stein A, Platzbecker U, Cross M. How Azanucleosides Affect Myeloid Cell Fate. Cells 2022; 11:cells11162589. [PMID: 36010665 PMCID: PMC9406747 DOI: 10.3390/cells11162589] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The azanucleosides decitabine and azacytidine are used widely in the treatment of myeloid neoplasia and increasingly in the context of combination therapies. Although they were long regarded as being largely interchangeable in their function as hypomethylating agents, the azanucleosides actually have different mechanisms of action; decitabine interferes primarily with the methylation of DNA and azacytidine with that of RNA. Here, we examine the role of DNA methylation in the lineage commitment of stem cells during normal hematopoiesis and consider how mutations in epigenetic regulators such as DNMT3A and TET2 can lead to clonal expansion and subsequent neoplastic progression. We also consider why the efficacy of azanucleoside treatment is not limited to neoplasias carrying mutations in epigenetic regulators. Finally, we summarise recent data describing a role for azacytidine-sensitive RNA methylation in lineage commitment and in the cellular response to stress. By summarising and interpreting evidence for azanucleoside involvement in a range of cellular processes, our review is intended to illustrate the need to consider multiple modes of action in the design and stratification of future combination therapies.
Collapse
|