1
|
Mediavilla C, D'Aveni M, Huynh A, Joris M, Masouridi-Levrat S, Nguyen S, Turlure P, Magro L, Guillaume T. [Place of hematopoietic stem cell transplantation for very high risk acute myeloblastic leukemia and myelodysplastic syndromes (SFGM-TC)]. Bull Cancer 2024:S0007-4551(24)00276-5. [PMID: 39242254 DOI: 10.1016/j.bulcan.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 09/09/2024]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloHSCT) is a potentially curative treatment for acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). However, these transplants are complicated by a high rate of relapse in very high cytogenetic risk or refractory diseases. The benefit of this therapeutic strategy for these serious malignant hemopathies could therefore be reassessed. As part of the 14th workshop for the harmonization of allograft practices organized by the francophone society of bone marrow transplantation and cellular therapy (SFGM-TC) (SFGM-TC) in Lille in September 2023, the role of allograft for very high risk or refractory AML and MDS was challenged after analysis of published studies.
Collapse
Affiliation(s)
| | - Maud D'Aveni
- Service d'hématologie, CHU de Nancy, Nancy, France
| | - Anne Huynh
- Service d'hématologie, IUCT, CHU de Toulouse, Toulouse, France
| | | | | | - Stéphanie Nguyen
- Service d'hématologie, CHU La Pitié-Salpétrière, Sorbonne Université, Paris, France
| | - Pascal Turlure
- Service d'hématologie, CHU Dupuytren Limoges, 2, avenue Martin Luther King, 87000 Limoges, France
| | - Leonardo Magro
- Service des maladies du sang, CHU de Lille, 2, rue Michel Polonowski, 59000 Lille, France
| | - Thierry Guillaume
- Service d'hématologie clinique, CHU de Nantes, Hôtel Dieu, 1, place Ricordeau, 44000 Nantes, France.
| |
Collapse
|
2
|
Silva-Hurtado TJ, Inocencio JF, Yong RL. Emerging applications of hypomethylating agents in the treatment of glioblastoma (Review). Mol Clin Oncol 2024; 21:59. [PMID: 39006906 PMCID: PMC11240870 DOI: 10.3892/mco.2024.2757] [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: 03/08/2024] [Accepted: 04/26/2024] [Indexed: 07/16/2024] Open
Abstract
DNA hypomethylating agents (HMAs) such as decitabine and 5-azacytidine have established roles in the treatment paradigms for myelodysplastic syndrome and acute myelogenous leukemia, where they are considered to exert their anticancer effects by restoring the expression of tumor suppressor genes. Due to their relatively favorable adverse effect profile and known ability to pass through the blood-brain barrier, applications in the treatment of glioblastoma (GBM) and other central nervous system malignancies are under active investigation. The present review examines the types of HMAs currently available, their known and less-understood antineoplastic mechanisms, and the evidence to date of their preclinical and clinical efficacy in glioblastoma and other solid malignancies. The present review discusses the potential synergies HMAs may have with established and emerging GBM treatments, including temozolomide, immune checkpoint inhibitors and cancer vaccines. Recent successes and setbacks in clinical trials for newly diagnosed and recurrent GBM are summarized in order to highlight opportunities for HMAs to improve therapeutic responses. Challenges for future clinical trials are also assessed.
Collapse
Affiliation(s)
- Thenzing J. Silva-Hurtado
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Julio F. Inocencio
- Leo M. Davidoff Department of Neurosurgery, Montefiore Medical Center, Einstein College of Medicine, Bronx, NY 10461, USA
| | - Raymund L. Yong
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
3
|
Sampaio LR, Dias RDB, Goes JVC, de Melo RPM, de Paula Borges D, de Lima Melo MM, de Oliveira RTG, Ribeiro-Júnior HL, Magalhães SMM, Pinheiro RF. Role of the STING pathway in myeloid neoplasms: a prospero-registered systematic review of principal hurdles of STING on the road to the clinical practice. Med Oncol 2024; 41:128. [PMID: 38656461 DOI: 10.1007/s12032-024-02376-8] [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: 03/10/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024]
Abstract
Myeloid neoplasms are a group of bone marrow diseases distinguished by disruptions in the molecular pathways that regulate the balance between hematopoietic stem cell (HSC) self-renewal and the generation of specialized cells. Cytokines and chemokines, two important components of the inflammatory process, also influence hematological differentiation. In this scenario, immunological dysregulation plays a pivotal role in the pathogenesis of bone marrow neoplasms. The STING pathway recognizes DNA fragments in the cell cytoplasm and triggers an immune response by type I interferons. The role of STING in cancer has not yet been established; however, both actions, as an oncogene or tumor suppressor, have been documented in other types of cancer. Therefore, we performed a systematic review (registered in PROSPERO database #CRD42023407512) to discuss the role of STING pathway in the advancement of pathogenesis and/or prognosis for different myeloid neoplasms. In brief, scientific evidence supports investigations that primarily use cell lines from myeloid neoplasms, such as leukemia. More high-quality research and clinical trials are needed to understand the role of the STING pathway in the pathology of hematological malignancies. Finally, the STING pathway suggests being a promising therapeutic molecular target, particularly when combined with current drug therapies.
Collapse
Affiliation(s)
- Leticia Rodrigues Sampaio
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Ricardo Dyllan Barbosa Dias
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - João Vitor Caetano Goes
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Renata Pinheiro Martins de Melo
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Daniela de Paula Borges
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Mayara Magna de Lima Melo
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Roberta Taiane Germano de Oliveira
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Howard Lopes Ribeiro-Júnior
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Silvia Maria Meira Magalhães
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil
| | - Ronald Feitosa Pinheiro
- Cancer Cytogenomic Laboratory, Federal University of Ceara, Fortaleza, Ceara, Brazil.
- Post-Graduate Program in Medical Science, Federal University of Ceara, Fortaleza, Ceara, Brazil.
- Drug Research and Development Center (NPDM), Federal University of Ceara, Fortaleza, Ceara, Brazil.
- Post-Graduate Program of Pathology, Federal University of Ceara, Fortaleza, Ceara, Brazil.
| |
Collapse
|
4
|
Janssen LL, Westers TM, Rovers J, Valk PJ, Cloos J, de Gruijl TD, van de Loosdrecht AA. Durable Responses and Survival in High-risk Myelodysplastic Syndrome and Acute Myeloid Leukemia Patients Receiving the Allogeneic Leukemia-derived Dendritic Cell Vaccine DCP-001. Hemasphere 2023; 7:e968. [PMID: 37928626 PMCID: PMC10624465 DOI: 10.1097/hs9.0000000000000968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023] Open
Affiliation(s)
- Luca L.G. Janssen
- Amsterdam UMC, Vrije Universiteit, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Theresia M. Westers
- Amsterdam UMC, Vrije Universiteit, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | - Peter J.M. Valk
- Erasmus University Medical Center, Department of Hematology, Rotterdam, The Netherlands
| | - Jacqueline Cloos
- Amsterdam UMC, Vrije Universiteit, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Tanja D. de Gruijl
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam UMC, Vrije Universiteit, Department of Medical Oncology, Amsterdam, The Netherlands
| | - Arjan A. van de Loosdrecht
- Amsterdam UMC, Vrije Universiteit, Department of Hematology, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Costa B, Vale N. Exploring HERV-K (HML-2) Influence in Cancer and Prospects for Therapeutic Interventions. Int J Mol Sci 2023; 24:14631. [PMID: 37834078 PMCID: PMC10572383 DOI: 10.3390/ijms241914631] [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: 08/29/2023] [Revised: 09/19/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
This review investigates the intricate role of human endogenous retroviruses (HERVs) in cancer development and progression, explicitly focusing on HERV-K (HML-2). This paper sheds light on the latest research advancements and potential treatment strategies by examining the historical context of HERVs and their involvement in critical biological processes such as embryonic development, immune response, and disease progression. This review covers computational modeling for drug-target binding assessment, systems biology modeling for simulating HERV-K viral cargo dynamics, and using antiviral drugs to combat HERV-induced diseases. The findings presented in this review contribute to our understanding of HERV-mediated disease mechanisms and provide insights into future therapeutic approaches. They emphasize why HERV-K holds significant promise as a biomarker and a target.
Collapse
Affiliation(s)
- Bárbara Costa
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| |
Collapse
|
6
|
Penter L, Liu Y, Wolff JO, Yang L, Taing L, Jhaveri A, Southard J, Patel M, Cullen NM, Pfaff KL, Cieri N, Oliveira G, Kim-Schulze S, Ranasinghe S, Leonard R, Robertson T, Morgan EA, Chen HX, Song MH, Thurin M, Li S, Rodig SJ, Cibulskis C, Gabriel S, Bachireddy P, Ritz J, Streicher H, Neuberg DS, Hodi FS, Davids MS, Gnjatic S, Livak KJ, Altreuter J, Michor F, Soiffer RJ, Garcia JS, Wu CJ. Mechanisms of response and resistance to combined decitabine and ipilimumab for advanced myeloid disease. Blood 2023; 141:1817-1830. [PMID: 36706355 PMCID: PMC10122106 DOI: 10.1182/blood.2022018246] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/29/2023] Open
Abstract
The challenge of eradicating leukemia in patients with acute myelogenous leukemia (AML) after initial cytoreduction has motivated modern efforts to combine synergistic active modalities including immunotherapy. Recently, the ETCTN/CTEP 10026 study tested the combination of the DNA methyltransferase inhibitor decitabine together with the immune checkpoint inhibitor ipilimumab for AML/myelodysplastic syndrome (MDS) either after allogeneic hematopoietic stem cell transplantation (HSCT) or in the HSCT-naïve setting. Integrative transcriptome-based analysis of 304 961 individual marrow-infiltrating cells for 18 of 48 subjects treated on study revealed the strong association of response with a high baseline ratio of T to AML cells. Clinical responses were predominantly driven by decitabine-induced cytoreduction. Evidence of immune activation was only apparent after ipilimumab exposure, which altered CD4+ T-cell gene expression, in line with ongoing T-cell differentiation and increased frequency of marrow-infiltrating regulatory T cells. For post-HSCT samples, relapse could be attributed to insufficient clearing of malignant clones in progenitor cell populations. In contrast to AML/MDS bone marrow, the transcriptomes of leukemia cutis samples from patients with durable remission after ipilimumab monotherapy showed evidence of increased infiltration with antigen-experienced resident memory T cells and higher expression of CTLA-4 and FOXP3. Altogether, activity of combined decitabine and ipilimumab is impacted by cellular expression states within the microenvironmental niche of leukemic cells. The inadequate elimination of leukemic progenitors mandates urgent development of novel approaches for targeting these cell populations to generate long-lasting responses. This trial was registered at www.clinicaltrials.gov as #NCT02890329.
Collapse
Affiliation(s)
- Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yang Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Lin Yang
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Len Taing
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Aashna Jhaveri
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Jackson Southard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA
| | - Manishkumar Patel
- Human Immune Monitoring Center at the Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nicole M. Cullen
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Kathleen L. Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Nicoletta Cieri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center at the Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Rebecca Leonard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Taylor Robertson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Elizabeth A. Morgan
- Harvard Medical School, Boston, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Helen X. Chen
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Minkyung H. Song
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Magdalena Thurin
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA
| | - Scott J. Rodig
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Carrie Cibulskis
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Stacey Gabriel
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | | | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Howard Streicher
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Donna S. Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sacha Gnjatic
- Human Immune Monitoring Center at the Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kenneth J. Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA
| | | | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Robert J. Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
7
|
Serroukh Y, Hébert J, Busque L, Mercier F, Rudd CE, Assouline S, Lachance S, Delisle JS. Blasts in context: the impact of the immune environment on acute myeloid leukemia prognosis and treatment. Blood Rev 2023; 57:100991. [PMID: 35941029 DOI: 10.1016/j.blre.2022.100991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a cancer that originates from the bone marrow (BM). Under physiological conditions, the bone marrow supports the homeostasis of immune cells and hosts memory lymphoid cells. In this review, we summarize our present understanding of the role of the immune microenvironment on healthy bone marrow and on the development of AML, with a focus on T cells and other lymphoid cells. The types and function of different immune cells involved in the AML microenvironment as well as their putative role in the onset of disease and response to treatment are presented. We also describe how the immune context predicts the response to immunotherapy in AML and how these therapies modulate the immune status of the bone marrow. Finally, we focus on allogeneic stem cell transplantation and summarize the current understanding of the immune environment in the post-transplant bone marrow, the factors associated with immune escape and relevant strategies to prevent and treat relapse.
Collapse
Affiliation(s)
- Yasmina Serroukh
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Erasmus Medical center Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada.
| | - Josée Hébert
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada; The Quebec Leukemia Cell Bank, Canada
| | - Lambert Busque
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - François Mercier
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Christopher E Rudd
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Sarit Assouline
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Silvy Lachance
- Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Jean-Sébastien Delisle
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| |
Collapse
|
8
|
Kim M, Delgado E, Ko S. DNA methylation in cell plasticity and malignant transformation in liver diseases. Pharmacol Ther 2023; 241:108334. [PMID: 36535346 PMCID: PMC9841769 DOI: 10.1016/j.pharmthera.2022.108334] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
The liver possesses extraordinary regenerative capacity mainly attributable to the ability of hepatocytes (HCs) and biliary epithelial cells (BECs) to self-replicate. This ability is left over from their bipotent parent cell, the hepatoblast, during development. When this innate regeneration is compromised due to the absence of proliferative parenchymal cells, such as during cirrhosis, HCs and BEC can transdifferentiate; thus, adding another layer of complexity to the process of liver repair. In addition, dysregulated lineage maintenance in these two cell populations has been shown to promote malignant growth in experimental conditions. Here, malignant transformation, driven in part by insufficient maintenance of lineage reprogramming, contributes to end-stage liver disease. Epigenetic changes are key drivers for cell fate decisions as well as transformation by finetuning overall transcription and gene expression. In this review, we address how altered DNA methylation contributes to the initiation and progression of hepatic cell fate conversion and cancer formation. We also discussed the diagnostic and therapeutic potential of targeting DNA methylation in liver cancer, its current limitations, and what future research is necessary to facilitate its contribution to clinical translation.
Collapse
Affiliation(s)
- Minwook Kim
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Evan Delgado
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Sungjin Ko
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America.
| |
Collapse
|
9
|
Nayak D, Weadick B, Govindarajan R. Combination of Tissue Microarray Profiling and Multiplexed IHC Approaches to Investigate Transport Mechanism of Nucleoside Analog Drug Resistance. Methods Mol Biol 2023; 2660:95-121. [PMID: 37191793 PMCID: PMC10311792 DOI: 10.1007/978-1-0716-3163-8_8] [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] [Indexed: 05/17/2023]
Abstract
Nucleoside analogs (NAs) are an established class of anticancer agents being used clinically for the treatment of diverse cancers, either as monotherapy or in combination with other established anticancer or pharmacological agents. To date, nearly a dozen anticancer NAs are approved by the FDA, and several novel NAs are being tested in preclinical and clinical trials for future applications. However, improper delivery of NAs into tumor cells because of alterations in expression of one or more drug carrier proteins (e.g., solute carrier (SLC) transporters) within tumor cells or cells surrounding the tumor microenvironment stands as one of the primary reasons for therapeutic drug resistance. The combination of tissue microarray (TMA) and multiplexed immunohistochemistry (IHC) is an advanced, high-throughput approach over conventional IHC that enables researchers to effectively investigate alterations to numerous such chemosensitivity determinants simultaneously in hundreds of tumor tissues derived from patients. In this chapter, taking an example of a TMA from pancreatic cancer patients treated with gemcitabine (a NA chemotherapeutic agent), we describe the step-by-step procedure of performing multiplexed IHC, imaging of TMA slides, and quantification of expression of some relevant markers in these tissue sections as optimized in our laboratory and discuss considerations while designing and carrying out this experiment.
Collapse
Affiliation(s)
- Debasis Nayak
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - Brenna Weadick
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, USA.
- Translational Therapeutics, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
10
|
Upadhyay P, Beales J, Shah NM, Gruszczynska A, Miller CA, Petti AA, Ramakrishnan SM, Link DC, Ley TJ, Welch JS. Recurrent transcriptional responses in AML and MDS patients treated with decitabine. Exp Hematol 2022; 111:50-65. [PMID: 35429619 PMCID: PMC9833843 DOI: 10.1016/j.exphem.2022.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 01/21/2023]
Abstract
The molecular events responsible for decitabine responses in myelodysplastic syndrome and acute myeloid leukemia patients are poorly understood. Decitabine has a short serum half-life and limited stability in tissue culture. Therefore, theoretical pharmacologic differences may exist between patient molecular changes in vitro and the consequences of in vivo treatment. To systematically identify the global genomic and transcriptomic alterations induced by decitabine in vivo, we evaluated primary bone marrow samples that were collected during patient treatment and applied whole-genome bisulfite sequencing, RNA-sequencing, and single-cell RNA sequencing. Decitabine induced global, reversible hypomethylation after 10 days of therapy in all patients, which was associated with induction of interferon-induced pathways, the expression of endogenous retroviral elements, and inhibition of erythroid-related transcripts, recapitulating many effects seen previously in in vitro studies. However, at relapse after decitabine treatment, interferon-induced transcripts remained elevated relative to day 0, but erythroid-related transcripts now were more highly expressed than at day 0. Clinical responses were not correlated with epigenetic or transcriptional signatures, although sample size and interpatient variance restricted the statistical power required for capturing smaller effects. Collectively, these data define global hypomethylation by decitabine and find that erythroid-related pathways may be relevant because they are inhibited by therapy and reverse at relapse.
Collapse
Affiliation(s)
- Pawan Upadhyay
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jeremy Beales
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Nakul M. Shah
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Agata Gruszczynska
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Christopher A. Miller
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Allegra A. Petti
- Department of Neuro-logical Surgery, Washington University School of Medicine, St. Louis, MO
| | - Sai Mukund Ramakrishnan
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Daniel C. Link
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Timothy J. Ley
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - John S. Welch
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
11
|
Kotnova AP, Stepchenko AG, Ilyin YV, Georgieva SG, Pankratova EV. 5-Azacytidine Suppresses the Expression of Tissue-Specific Oct-1 Isoform in Namalwa Burkitt's Lymphoma Cell Culture. DOKL BIOCHEM BIOPHYS 2022; 503:76-79. [PMID: 35538282 PMCID: PMC9090699 DOI: 10.1134/s1607672922020089] [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: 11/30/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 11/25/2022]
Abstract
Overexpression of the transcription factor POU2F1 (Oct-1) increases the malignant potential of the tumor and determines the unfavorable prognosis for both solid and hematological cases of the disease in human carcinogenesis. The Oct-1 level determines the rate of development of the disease in acute myelodysplastic leukemia (AML), and a decrease in its expression significantly delays the development of leukemia in mice; however, a complete knockout of Oct-1 leads to the death of the animals. POU2F1 (Oct-1) is expressed as several isoforms transcribed from alternative promoters. They include both ubiquitous and tissue-specific isoforms. It was shown that in Burkitt’s lymphoma Namalwa cells 5-azacytidine specifically suppresses the expression of the tissue-specific isoform Oct-1L mRNA (level of Oct-1L is abnormally increased in these cells), while not causing changes in the amount of the ubiquitous isoform Oct-1A mRNA. These results show that it is possible to selectively reduce the transcription level of the Oct-1L isoform aberrantly expressed in human tumor cells.
Collapse
Affiliation(s)
- A P Kotnova
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia.
| | - A G Stepchenko
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
| | - Yu V Ilyin
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
| | - S G Georgieva
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
| | - E V Pankratova
- Engelhardt Institute of Molecular Biology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
12
|
Su N, Wang Y, Lu X, Xu W, Wang H, Mo W, Pang H, Tang R, Li S, Yan X, Li Y, Zhang R. Methylation of SPRED1: A New Target in Acute Myeloid Leukemia. Front Oncol 2022; 12:854192. [PMID: 35359401 PMCID: PMC8960233 DOI: 10.3389/fonc.2022.854192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/14/2022] [Indexed: 11/22/2022] Open
Abstract
Sprouty-related, EVH1 domain-containing protein 1 (SPRED1) has been identified as a novel tumor suppressor gene in acute myeloid leukemia (AML). Previous studies showed that SPRED1 methylation levels were significantly increased in AML patients, making it an interesting candidate for further investigations. To confirm the association of SPRED1 methylation, clinical parameters, and known molecular prognosticators and to identify the impact of methylation level on treatment outcome, we conducted this study in a larger cohort of 75 AML patients. Significantly increased methylation levels of SPRED1 were detected at four of ten CpG units by quantitative high-resolution mass spectrometry-based approach (MassARRAY) in AML patients. Whereas overall survival (OS) and relapse-free survival (RFS) showed no statistical difference between hypermethylation and hypomethylation subgroups, the relationship between methylation level and treatment response was indicated in paired samples from pre- and post-induction. To determine the possible mechanism of SPRED1 methylation in AML, we performed in vitro experiments using THP-1 cells, as the latter showed the highest methylation level (determined by utilizing bisulfite modification) among the three AML cell lines we tested. When treated with 5-AZA and lentivirus transfection, upregulated SPRED1 expression, decreased cell proliferation, increased cell differentiation and apoptosis, and inactivated phosphorylated extracellular signal-regulated kinase (p-ERK) were detected in THP-1 cells. These results show that demethylation of SPRED1 can inhibit the proliferation of AML cells and promote their differentiation and apoptosis, possibly by the ERK pathway. The hypermethylation of SPRED1 is a potential therapeutic target for AML.
Collapse
Affiliation(s)
- Nan Su
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yujiao Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xianglan Lu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Weihong Xu
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - He Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Wenbin Mo
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Pang
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rurong Tang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shibo Li
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xiaojing Yan
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Li
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Rui Zhang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Metronomic chemotherapy regimens and targeted therapies in non-Hodgkin lymphoma: The best of two worlds. Cancer Lett 2022; 524:144-150. [PMID: 34673128 DOI: 10.1016/j.canlet.2021.10.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/31/2022]
Abstract
Novel drugs are rapidly moving forward the treatment-paradigm of non-Hodgkin-lymphomas (NHLs). Notwithstanding, especially in aggressive subtypes, chemotherapy remains the pillar of treatment. Indeed, the combination of highly effective Maximum-Tolerated-Dose Chemotherapy (MTD-CHEMO) + "novel drugs", has so far, fallen short from expectations, often because it caused excessive toxicity. Metronomic chemotherapy (mCHEMO), which is the frequent, long-term administration of low dose cytotoxic drugs, may allow more effective and tolerable combinations. mCHEMO pharmacodynamics, has been described as pleiotropic. In fact, it may have different cellular and molecular targets, when drugs or their schedules are modified. Although mCHEMO has been little explored in NHLs, pre-clinical studies - in lymphoma models - which addressed the activity of mCHEMO in combination with novel drugs, have shown very promising results. These included inhibitors of histone deacetylase, mTOR and PI3K/mTOR, as well as the immune checkpoint inhibitor anti-PD-L1. Moreover, a few impressive reports have recently shown all-oral mCHEMO schedules, with or without rituximab, can effectively shrink both B and T-cell aggressive NHLs. Indeed, these regimens allowed elderly-frail patients to achieve sustained remission, while toxicity proved manageable. In our opinion, all-oral mCHEMO, is an active, easy-to start, well-tolerated, and inexpensive therapeutic approach, which deserves further investigation. Most importantly, mCHEMO, holds promise to empower the activity of novel targeted therapies, without causing excessive toxicity.
Collapse
|
14
|
Baroud M, Lepeltier E, El-Makhour Y, Lautram N, Bejaud J, Thepot S, Duval O. Azacitidine Omega-3 Self-Assemblies: Synthesis, Characterization, and Potent Applications for Myelodysplastic Syndromes. Pharmaceuticals (Basel) 2021; 14:1317. [PMID: 34959720 PMCID: PMC8706301 DOI: 10.3390/ph14121317] [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: 10/31/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 11/30/2022] Open
Abstract
5-Azacitidine, a cytidine analogue used as a hypomethylating agent, is one of the main drugs for the treatment of myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML) in the elderly. However, after administration, it exhibits several limitations, including restricted diffusion and cellular internalization due to its hydrophilicity, and a rapid enzymatic degradation by adenosine deaminase. The aim of this study was to improve the drug cell diffusion and protect it from metabolic degradation via the synthesis of amphiphilic prodrugs and their potential self-assembly. Azacitidine was conjugated to two different omega-3 fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). The carboxylic acid group of the omega-3 fatty acids was effectively conjugated to the amine group of the azacitidine base, yielding two amphiphilic prodrugs. Nanoprecipitation of the obtained prodrugs was performed and self-assemblies were successfully obtained for both prodrugs, with a mean diameter of 190 nm, a polydispersity index below 0.2 and a positive zeta potential. The formation of self-assemblies was confirmed using pyrene as a fluorescent dye, and the critical aggregation concentrations were determined: 400 µM for AzaEPA and 688 µM for AzaDHA. Additionally, the stability of the obtained self-assemblies was studied and after 5 days their final stable arrangement was reached. Additionally, cryo-TEM revealed that the self-assemblies attain a multilamellar vesicle supramolecular structure. Moreover, the obtained self-assemblies presented promising cytotoxicity on a leukemia human cell line, having a low IC50 value, comparable to that of free azacitidine.
Collapse
Affiliation(s)
- Milad Baroud
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Elise Lepeltier
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Yolla El-Makhour
- Environmental Health Research Lab, Faculty of Science, Lebanese University, Nabatieh 1700, Lebanon;
| | - Nolwenn Lautram
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Jerome Bejaud
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
| | - Sylvain Thepot
- Department of Hematology, University Hospital of Angers, 49933 Angers, France;
- Federation Hospital of Universitaire Grand Ouest Acute Leukemia (FHU GOAL), 49933 Angers, France
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERM, University of Angers, 49933 Angers, France
| | - Olivier Duval
- Micro & Nanomedecines Translationnelles (MINT), Inserm, The National Center for Scientific Research (CNRS), SFR ICAT, University of Angers, 49000 Angers, France; (M.B.); (E.L.); (N.L.); (J.B.)
- Department of Hematology, University Hospital of Angers, 49933 Angers, France;
| |
Collapse
|
15
|
Grauers Wiktorin H, Aydin E, Christenson K, Issdisai N, Thorén FB, Hellstrand K, Martner A. Impact of IL-1β and the IL-1R antagonist on relapse risk and survival in AML patients undergoing immunotherapy for remission maintenance. Oncoimmunology 2021; 10:1944538. [PMID: 34367728 PMCID: PMC8317920 DOI: 10.1080/2162402x.2021.1944538] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Interleukin-1 beta (IL-1β), a pro-inflammatory cytokine, has been ascribed a role in the expansion of myeloid progenitors in acute myeloid leukemia (AML) and in promoting myeloid cell-induced suppression of lymphocyte-mediated immunity against malignant cells. This study aimed at defining the potential impact of IL-1β in the post-remission phase of AML patients receiving immunotherapy for relapse prevention in an international phase IV trial of 84 patients (ClinicalTrials.gov; NCT01347996). Consecutive serum samples were collected from AML patients in first complete remission (CR) who received cycles of relapse-preventive immunotherapy with histamine dihydrochloride (HDC) and low-dose interleukin-2 (IL-2). Low IL-1β serum levels before and after the first HDC/IL-2 treatment cycle favorably prognosticated leukemia-free survival and overall survival. Serum levels of IL-1β were significantly reduced in patients receiving HDC/IL-2. HDC also reduced the formation of IL-1β from activated human PBMCs in vitro. Additionally, high serum levels of the IL-1 receptor antagonist IL-1RA were associated with favorable outcome, and AML patients with low IL-1β along with high IL-1RA levels were strikingly protected against leukemic relapse. Our results suggest that strategies to target IL-1β might impact on relapse risk and survival in AML.
Collapse
Affiliation(s)
- Hanna Grauers Wiktorin
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Ebru Aydin
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.,Molecular Genetics, Germany Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Christenson
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden.,Department of Oral Microbiology and Immunology, Institute of Odontology, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Nuttida Issdisai
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Fredrik B Thorén
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Kristoffer Hellstrand
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Center for Cancer Research, Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|