1
|
Zhang X, Zhong M, Fu X, Pan H, Liu H, Chen J, Guo F. Exploring the oncogenic potential of Aiolos in lung cancer through OTUB1-mediated ubiquitination. Heliyon 2024; 10:e37710. [PMID: 39315162 PMCID: PMC11417157 DOI: 10.1016/j.heliyon.2024.e37710] [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: 06/09/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Aiolos (IKZF3), a zinc finger transcription factor, has been identified in various solid tumors. While most research on Aiolos focuses on its role in the hematopoietic system, its expression patterns, mechanisms of action, and biological impacts in lung cancer remain relatively unexplored. This study investigates Aiolos' role in the proliferation, migration, and invasion of lung cancer cells. Our findings indicate that Aiolos overexpression enhances these cellular processes, suggesting its potential contribution to the advancement of the disease. However, the precise mechanisms underlying these effects require further investigation. Additionally, we identified OTUB1 as a potential Aiolos-interacting protein. OTUB1, a deubiquitinating enzyme, removes ubiquitin chains from target proteins, thereby affecting their stability, function, or localization. Our results suggest that OTUB1 specially bound to Aiolos and reduces its ubiquitination, which may influence Aiolos-related biological functions, including cell migration and invasion. This study highlights the pivotal roles of Aiolos and OTUB1 in lung cancer progression, potentially offering new therapeutic targets.
Collapse
Affiliation(s)
- Xiuwen Zhang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Mei Zhong
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xinyue Fu
- The South China University of Technology School of Medicine, Guangzhou, 510006, China
| | - Hongli Pan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jun Chen
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Fengjie Guo
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
- The South China University of Technology School of Medicine, Guangzhou, 510006, China
| |
Collapse
|
2
|
Tebbi CK, Yan J, Sahakian E, Mediavilla-Varela M, Pinilla-Ibarz J, Patel S, Rottinghaus GE, Liu RY, Dennison C. Mycovirus-Containing Aspergillus flavus Alters Transcription Factors in Normal and Acute Lymphoblastic Leukemia Cells. Int J Mol Sci 2024; 25:10361. [PMID: 39408690 PMCID: PMC11476453 DOI: 10.3390/ijms251910361] [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: 06/24/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/20/2024] Open
Abstract
Transcription factors control genes to maintain normal hemopoiesis, and dysregulation of some factors can lead to acute lymphoblastic leukemia (ALL). Mycoviruses are known to alter the genetics of their fungal host. The present study evaluates the effects of the products of a mycovirus-containing Aspergillus flavus (MCAF), isolated from the home of a patient with ALL, on certain transcription factors of normal and ALL cell lines. Our published studies have shown that ALL patients have antibodies to MCAF, and that exposure of the mononuclear leukocytes of patients in complete remission to its products, unlike controls, results in the re-development of genetic and cell surface phenotypes characteristic of ALL. For the present study, normal, pre-B, and B-cell leukemia cell lines were exposed to the culture of MCAF. Pre- and post-exposure levels of PAX5, Ikaros, and NF-κB were assessed. Exposure to MCAF resulted in apoptosis, cell cycle changes, and complete downregulation of all transcription factors in normal cell lines. In acute leukemia cell lines, cellular apoptosis and alterations in the cell cycle were also noted; however, while there was downregulation of all tested transcription factors, residual levels were retained. The noted alterations in the transcription factors caused by MCAF are novel findings. The possible role of MCAF in leukemogenesis needs to be further investigated. Mycovirus-containing Aspergillus flavus was initially isolated from a leukemia patient's home. Our prior published studies have illuminated intriguing associations of this organism with leukemia. Unlike controls, patients diagnosed with acute lymphoblastic leukemia (ALL) harbor antibodies to this organism. Furthermore, the exposure of mononuclear cells from patients with ALL in complete remission to the products of this organism reproduced genetic and cell phenotypes characteristic of ALL. These findings underscore the potential role of environmental factors in leukemogenesis and hint at novel avenues for therapeutic intervention and preventive strategies.
Collapse
Affiliation(s)
- Cameron K. Tebbi
- Children’s Cancer Research Group Laboratory, Tampa, FL 33613, USA; (J.Y.); (R.Y.L.)
| | - Jiyu Yan
- Children’s Cancer Research Group Laboratory, Tampa, FL 33613, USA; (J.Y.); (R.Y.L.)
| | - Eva Sahakian
- Moffitt Cancer Center, Tampa, FL 33612, USA; (E.S.); (M.M.-V.); (J.P.-I.)
| | | | | | | | | | - Rachel Y. Liu
- Children’s Cancer Research Group Laboratory, Tampa, FL 33613, USA; (J.Y.); (R.Y.L.)
| | - Clare Dennison
- Diagnostic Laboratories, College of Veterinary Medicine, University of South Florida, Tampa, FL 33620, USA;
| |
Collapse
|
3
|
Ichiyama K, Long J, Kobayashi Y, Horita Y, Kinoshita T, Nakamura Y, Kominami C, Georgopoulos K, Sakaguchi S. Transcription factor Ikzf1 associates with Foxp3 to repress gene expression in Treg cells and limit autoimmunity and anti-tumor immunity. Immunity 2024; 57:2043-2060.e10. [PMID: 39111316 DOI: 10.1016/j.immuni.2024.07.010] [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: 08/10/2023] [Revised: 02/16/2024] [Accepted: 07/15/2024] [Indexed: 09/13/2024]
Abstract
The master transcription factor of regulatory T (Treg) cells, forkhead box protein P3 (Foxp3), controls Treg cell function by targeting certain genes for activation or repression, but the specific mechanisms by which it mediates this activation or repression under different conditions remain unclear. We found that Ikzf1 associates with Foxp3 via its exon 5 (IkE5) and that IkE5-deficient Treg cells highly expressed genes that would otherwise be repressed by Foxp3 upon T cell receptor stimulation, including Ifng. Treg-specific IkE5-deletion caused interferon-γ (IFN-γ) overproduction, which destabilized Foxp3 expression and impaired Treg suppressive function, leading to systemic autoimmune disease and strong anti-tumor immunity. Pomalidomide, which degrades IKZF1 and IKZF3, induced IFN-γ overproduction in human Treg cells. Mechanistically, the Foxp3-Ikzf1-Ikzf3 complex competed with epigenetic co-activators, such as p300, for binding to target gene loci via chromatin remodeling. Therefore, the Ikzf1 association with Foxp3 is essential for the gene-repressive function of Foxp3 and could be exploited to treat autoimmune disease and cancer.
Collapse
Affiliation(s)
- Kenji Ichiyama
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan.
| | - Jia Long
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Yusuke Kobayashi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Medical Innovations, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Osaka, Japan
| | - Yuji Horita
- Joint Research Chair of Immune-therapeutic Drug Discovery, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Research Management, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Takeshi Kinoshita
- Joint Research Chair of Immune-therapeutic Drug Discovery, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Research Management, Otsuka Pharmaceutical Co., Ltd., Tokushima, Japan
| | - Yamami Nakamura
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Chizuko Kominami
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Katia Georgopoulos
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Shimon Sakaguchi
- Laboratory of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Experimental Pathology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan.
| |
Collapse
|
4
|
Tran NT, Graf R, Acevedo-Ochoa E, Trombke J, Weber T, Sommermann T, Salomon C, Kühn R, Rajewsky K, Chu VT. In vivo CRISPR/Cas9-mediated screen reveals a critical function of TFDP1 and E2F4 transcription factors in hematopoiesis. Leukemia 2024; 38:2003-2015. [PMID: 39043964 PMCID: PMC11347378 DOI: 10.1038/s41375-024-02357-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
Hematopoiesis is a continuous process of blood cell production driven by hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. Proliferation and differentiation of HSPCs are regulated by complex transcriptional networks. In order to identify transcription factors with key roles in HSPC-mediated hematopoietic reconstitution, we developed an efficient and robust CRISPR/Cas9-based in vivo genetic screen. Using this experimental system, we identified the TFDP1 transcription factor to be essential for HSPC proliferation and post-transplant hematopoiesis. We further discovered that E2F4, an E2F transcription factor, serves as a binding partner of TFDP1 and is required for HSPC proliferation. Deletion of TFDP1 caused downregulation of genes associated with the cell cycle, with around 50% of these genes being identified as direct targets of TFDP1 and E2F4. Thus, our study expands the transcriptional network governing hematopoietic development through an in vivo CRISPR/Cas9-based genetic screen and identifies TFDP1/E2F4 as positive regulators of cell cycle genes in HSPCs.
Collapse
Affiliation(s)
- Ngoc Tung Tran
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany.
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Robin Graf
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, 13125, Germany
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin, Berlin, Germany
| | - Ernesto Acevedo-Ochoa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany
| | - Janine Trombke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany
| | - Timm Weber
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany
- Biobank OWL (BOWL), Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Thomas Sommermann
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany
- Dynamic42 GmbH, Jena, Germany
| | - Claudia Salomon
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany
| | - Ralf Kühn
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Genome Engineering & Disease Models, Berlin, Germany
| | - Klaus Rajewsky
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany.
| | - Van Trung Chu
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Immune Regulation and Cancer, Berlin, Germany.
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Genome Engineering & Disease Models, Berlin, Germany.
| |
Collapse
|
5
|
Østergaard A, Boer JM, van Leeuwen FN, Pieters R, Den Boer ML. IKZF1 in acute lymphoblastic leukemia: the rise before the fall? Leuk Lymphoma 2024:1-11. [PMID: 39210599 DOI: 10.1080/10428194.2024.2396046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in children and adolescents and in recent decades, the survival rates have risen to >90% in children largely due the introduction of risk adapted therapy. Therefore, knowledge of factors influencing risk of relapse is important. The transcription factor IKAROS is a regulator of lymphocyte development and alterations of its coding gene, IKZF1, are frequent in ALL and are associated with higher relapse risk. This concise review will discuss the normal function of IKAROS together with the effect of gene alterations in ALL such as relieved energy restriction and altered response to anti-leukemic drugs. Besides the biology, the clinical impact of gene alterations in the different subtypes of ALL will be discussed. Finally, possibilities for treating ALL with IKZF1 alterations will be considered including novel therapies like cell signaling inhibitors and immunotherapy.
Collapse
Affiliation(s)
- Anna Østergaard
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Judith M Boer
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | |
Collapse
|
6
|
Feng Y, Hu X, Wang X. Targeted protein degradation in hematologic malignancies: clinical progression towards novel therapeutics. Biomark Res 2024; 12:85. [PMID: 39169396 PMCID: PMC11340087 DOI: 10.1186/s40364-024-00638-1] [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: 06/30/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
Targeted therapies, such as small molecule kinase inhibitors, have made significant progress in the treatment of hematologic malignancies by directly modulating protein activity. However, issues such as drug toxicity, drug resistance due to target mutations, and the absence of key active sites limit the therapeutic efficacy of these drugs. Targeted protein degradation (TPD) presents an emergent and rapidly evolving therapeutic approach that selectively targets proteins of interest (POI) based on endogenous degradation processes. With an event-driven pharmacology of action, TPD achieves efficacy with catalytic amounts, avoiding drug-related toxicity. Furthermore, TPD has the unique mode of degrading the entire POI, such that resistance derived from mutations in the targeted protein has less impact on its degradation function. Proteolysis-targeting chimeras (PROTACs) and molecular glue degraders (MGDs) are the most maturely developed TPD techniques. In this review, we focus on both preclinical experiments and clinical trials to provide a comprehensive summary of the safety and clinical effectiveness of PROTACs and MGDs in hematologic malignancies over the past two decades. In addition, we also delineate the challenges and opportunities associated with these burgeoning degradation techniques. TPD, as an approach to the precise degradation of specific proteins, provides an important impetus for its future application in the treatment of patients with hematologic malignancies.
Collapse
Affiliation(s)
- Yupiao Feng
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|
7
|
Li M. IKZF2 Degradation: It's Time to Take into Account it When Designing Cereblon-Based PROTACs. Chembiochem 2024; 25:e202400365. [PMID: 38802326 DOI: 10.1002/cbic.202400365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 05/29/2024]
Abstract
Proteolysis-targeting chimera (PROTAC) has become a very important means of protein degradation and a new way of disease treatment. In particular, PROTACs constructed with ligands for E3 ligase cereblon account for more than 90 % of the PROTACs currently in clinical research. Notably, CRBN ligands themselves are a class of molecular glue compounds capable of degrading neo-substrate proteins. Compared to the target proteins degradation, the degradation of neo-substrates, especially IKZF2, has not received enough attention. Therefore, this review summarizes the currently published IKZF2 degraders derived from articles and patents, which are conducive to the design of PROTACs with desired IKZF2 degradation from the perspective of medicinal chemistry.
Collapse
Affiliation(s)
- Minglei Li
- Chemical Biology Center, School of Pharmaceutical Sciences & Institute of Materia Medical, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
- School of Pharmaceutical Sciences & Institute of Materia Medical, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, 250117, Shandong, China
| |
Collapse
|
8
|
Brodermann MH, Henderson EK, Sellar RS. The emerging role of targeted protein degradation to treat and study cancer. J Pathol 2024; 263:403-417. [PMID: 38886898 DOI: 10.1002/path.6301] [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: 01/18/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
The evolution of cancer treatment has provided increasingly targeted strategies both in the upfront and relapsed disease settings. Small-molecule inhibitors and immunotherapy have risen to prominence with chimeric antigen receptor T-cells, checkpoint inhibitors, kinase inhibitors, and monoclonal antibody therapies being deployed across a range of solid organ and haematological malignancies. However, novel approaches are required to target transcription factors and oncogenic fusion proteins that are central to cancer biology and have generally eluded successful drug development. Thalidomide analogues causing protein degradation have been a cornerstone of treatment in multiple myeloma, but a lack of in-depth mechanistic understanding initially limited progress in the field. When the protein cereblon (CRBN) was found to mediate thalidomide analogues' action and CRBN's neo-targets were identified, existing and novel drug development accelerated, with applications outside multiple myeloma, including non-Hodgkin's lymphoma, myelodysplastic syndrome, and acute leukaemias. Critically, transcription factors were the first canonical targets described. In addition to broadening the application of protein-degrading drugs, resistance mechanisms are being overcome and targeted protein degradation is widening the scope of druggable proteins against which existing approaches have been ineffective. Examples of targeted protein degraders include molecular glues and proteolysis targeting chimeras (PROTACs): heterobifunctional molecules that bind to proteins of interest and cause proximity-induced ubiquitination and proteasomal degradation via a linked E3 ligase. Twenty years since their inception, PROTACs have begun progressing through clinical trials, with early success in targeting the oestrogen receptor and androgen receptor in breast and prostate cancer respectively. This review explores important developments in targeted protein degradation to both treat and study cancer. It also considers the potential advantages and challenges in the translational aspects of developing new treatments. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Elizabeth K Henderson
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Rob S Sellar
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
9
|
Liu Y, Mo CC, Hartley-Brown MA, Sperling AS, Midha S, Yee AJ, Bianchi G, Piper C, Tattersall A, Nadeem O, Laubach JP, Richardson PG. Targeting Ikaros and Aiolos: reviewing novel protein degraders for the treatment of multiple myeloma, with a focus on iberdomide and mezigdomide. Expert Rev Hematol 2024; 17:445-465. [PMID: 39054911 DOI: 10.1080/17474086.2024.2382897] [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: 03/21/2024] [Revised: 05/30/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
INTRODUCTION The treatment of multiple myeloma (MM) is evolving rapidly. Quadruplet regimens incorporating proteasome inhibitors, immunomodulatory drugs (IMiDs), and CD38 monoclonal antibodies have emerged as standard-of-care options for newly diagnosed MM, and numerous novel therapies have been approved for relapsed/refractory MM. However, there remains a need for novel options in multiple settings, including refractoriness to frontline standards of care. AREAS COVERED Targeting degradation of IKZF1 and IKZF3 - Ikaros and Aiolos - through modulation of cereblon, an E3 ligase substrate recruiter/receptor, is a key mechanism of action of the IMiDs and the CELMoD agents. Two CELMoD agents, iberdomide and mezigdomide, have demonstrated substantial preclinical and clinical activity in MM and have entered phase 3 investigation. Using a literature search methodology comprising searches of PubMed (unlimited time-frame) and international hematology/oncology conference abstracts (2019-2023), this paper reviews the importance of Ikaros and Aiolos in MM, the mechanism of action of the IMiDs and CELMoD agents and their relative potency for targeting Ikaros and Aiolos, and preclinical and clinical data on iberdomide and mezigdomide. EXPERT OPINION Emerging data suggest that iberdomide and mezigdomide have promising activity, including in IMiD-resistant settings and, pending phase 3 findings, may provide additional treatment options for patients with MM.
Collapse
Affiliation(s)
- Yuxin Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Clifton C Mo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Monique A Hartley-Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Adam S Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shonali Midha
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Andrew J Yee
- Massachusetts General Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Giada Bianchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, USA
| | - Catherine Piper
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Alice Tattersall
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Omar Nadeem
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Jacob P Laubach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Van Oekelen O, Amatangelo M, Guo M, Upadhyaya B, Cribbs AP, Kelly G, Patel M, Kim-Schulze S, Flynt E, Lagana A, Gooding S, Merad M, Jagganath S, Pierceall WE, Oppermann U, Thakurta A, Parekh S. Iberdomide increases innate and adaptive immune cell subsets in the bone marrow of patients with relapsed/refractory multiple myeloma. Cell Rep Med 2024; 5:101584. [PMID: 38776911 PMCID: PMC11228551 DOI: 10.1016/j.xcrm.2024.101584] [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: 08/24/2023] [Revised: 02/11/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Iberdomide is a potent cereblon E3 ligase modulator (CELMoD agent) with promising efficacy and safety as a monotherapy or in combination with other therapies in patients with relapsed/refractory multiple myeloma (RRMM). Using a custom mass cytometry panel designed for large-scale immunophenotyping of the bone marrow tumor microenvironment (TME), we demonstrate significant increases of effector T and natural killer (NK) cells in a cohort of 93 patients with multiple myeloma (MM) treated with iberdomide, correlating findings to disease characteristics, prior therapy, and a peripheral blood immune phenotype. Notably, changes are dose dependent, associated with objective response, and independent of prior refractoriness to MM therapies. This suggests that iberdomide broadly induces innate and adaptive immune activation in the TME, contributing to its antitumor efficacy. Our approach establishes a strategy to study treatment-induced changes in the TME of patients with MM and, more broadly, patients with cancer and establishes rational combination strategies for iberdomide with immune-enhancing therapies to treat MM.
Collapse
Affiliation(s)
- Oliver Van Oekelen
- Department of Medicine, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Manman Guo
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Bhaskar Upadhyaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam P Cribbs
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Geoffrey Kelly
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manishkumar Patel
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erin Flynt
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, USA
| | - Alessandro Lagana
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Gooding
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Miriam Merad
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sundar Jagganath
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Udo Oppermann
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK; Oxford Translational Myeloma Centre (OTMC), Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Anjan Thakurta
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, USA; Oxford Translational Myeloma Centre (OTMC), Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Samir Parekh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
11
|
Shi X, Cao X, Huang M, Zhang P, Yang G, Ren A, Dai X, Chen R, Yang Z, Cai Z, Chen Y, Zhao X, Huang P, Du Z. Identification and Functional Analysis of a de novo IKZF3 Mutation in a Pediatric Patient with Combined Immunodeficiency. J Clin Immunol 2024; 44:117. [PMID: 38758229 DOI: 10.1007/s10875-024-01706-9] [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: 09/25/2023] [Accepted: 04/03/2024] [Indexed: 05/18/2024]
Abstract
AIOLOS, a vital member of the IKAROS protein family, plays a significant role in lymphocyte development and function through DNA binding and protein-protein interactions. Mutations in the IKZF3 gene, which encodes AIOLOS, lead to a rare combined immunodeficiency often linked with infections and malignancy. In this study, we evaluated a 1-year-4-month-old female patient presenting with recurrent infections, diarrhea, and failure to thrive. Laboratory investigations revealed decreased T lymphocyte and immunoglobulin levels. Through whole-exome and Sanger sequencing, we discovered a de novo mutation in IKZF3 (NM_012481; exon 5 c.571G > C, p.Gly191Arg), corresponding to the third DNA-binding zinc finger region of the encoded protein AIOLOS. Notably, the patient with the AIOLOS G191R mutation showed reduced recent thymic emigrants in naïve CD4+T cells compared to healthy counterparts of the same age, while maintaining normal levels of Th1, Th2, Th17, Treg, and Tfh cells. This mutation also resulted in decreased switched memory B cells and lower CD23 and IgM expression. In vitro studies revealed that AIOLOS G191R does not impact the expression of AIOLOS but compromises its stability, DNA binding and pericentromeric targeting. Furthermore, AIOLOS G191R demonstrated a dominant-negative effect over the wild-type protein. This case represents the first reported instance of a mutation in the third DNA-binding zinc finger region of AIOLOS highlighting its pivotal role in immune cell functionality.
Collapse
Affiliation(s)
- Xiaoqi Shi
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Xiuli Cao
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Meiying Huang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Pingping Zhang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Guangli Yang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Aiyan Ren
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Xin Dai
- Zhanjiang Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Department of Hematology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Ran Chen
- National Clinical Research Center for Child Health and Disorders (Chongqing), Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Yang
- Zhanjiang Institute of Clinical Medicine, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China
- Department of Hematology, Central People's Hospital of Zhanjiang, Zhanjiang, China
| | - Zeyuan Cai
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| | - Xiaodong Zhao
- National Clinical Research Center for Child Health and Disorders (Chongqing), Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Pei Huang
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China.
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China.
| | - Zuochen Du
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China.
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China.
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
12
|
Yu X, Chen Y, Chen J, Fan Y, Lu H, Wu D, Xu Y. Shared genetic architecture between autoimmune disorders and B-cell acute lymphoblastic leukemia: insights from large-scale genome-wide cross-trait analysis. BMC Med 2024; 22:161. [PMID: 38616254 PMCID: PMC11017616 DOI: 10.1186/s12916-024-03385-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND To study the shared genetic structure between autoimmune diseases and B-cell acute lymphoblastic leukemia (B-ALL) and identify the shared risk loci and genes and genetic mechanisms involved. METHODS Based on large-scale genome-wide association study (GWAS) summary-level data sets, we observed genetic overlaps between autoimmune diseases and B-ALL, and cross-trait pleiotropic analysis was performed to detect shared pleiotropic loci and genes. A series of functional annotation and tissue-specific analysis were performed to determine the influence of pleiotropic genes. The heritability enrichment analysis was used to detect crucial immune cells and tissues. Finally, bidirectional Mendelian randomization (MR) methods were utilized to investigate the casual associations. RESULTS Our research highlighted shared genetic mechanisms between seven autoimmune disorders and B-ALL. A total of 73 pleiotropic loci were identified at the genome-wide significance level (P < 5 × 10-8), 16 of which had strong evidence of colocalization. We demonstrated that several loci have been previously reported (e.g., 17q21) and discovered some novel loci (e.g., 10p12, 5p13). Further gene-level identified 194 unique pleiotropic genes, for example IKZF1, GATA3, IKZF3, GSDMB, and ORMDL3. Pathway analysis determined the key role of cellular response to cytokine stimulus, B cell activation, and JAK-STAT signaling pathways. SNP-level and gene-level tissue enrichment suggested that crucial role pleiotropic mechanisms involved in the spleen, whole blood, and EBV-transformed lymphocytes. Also, hyprcoloc and stratified LD score regression analyses revealed that B cells at different developmental stages may be involved in mechanisms shared between two different diseases. Finally, two-sample MR analysis determined causal effects of asthma and rheumatoid arthritis on B-ALL. CONCLUSIONS Our research proved shared genetic architecture between autoimmune disorders and B-ALL and shed light on the potential mechanism that might involve in.
Collapse
Affiliation(s)
- Xinghao Yu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Yiyin Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Jia Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Fan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huimin Lu
- Department of Outpatient and Emergency, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Collaborative Innovation Center of Hematology, Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China.
| |
Collapse
|
13
|
Singh V, Singh R, Kushwaha R. Exploring novel protein biomarkers for early-stage diagnosis and prognosis of T-acute lymphoblastic leukemia (T-ALL). Hematol Transfus Cell Ther 2024:S2531-1379(24)00063-4. [PMID: 38584071 DOI: 10.1016/j.htct.2024.02.016] [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: 08/26/2023] [Accepted: 02/12/2024] [Indexed: 04/09/2024] Open
Abstract
BACKGROUND Efficient classification of T-acute lymphoblastic leukemia (T-ALL) involves considering various factors, such as age, white blood cell count, and chromosomal alterations. However, studying protein markers are crucial to improving T-ALL patients' diagnosis and treatment. A study analyzing the expression of proteomes was conducted to identify promising early-stage biomarkers for T-ALL patients METHODS: Label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to analyze the blood proteins of both patients and healthy individuals to identify new biomarkers for T-ALL. The findings were validated by RT-PCR, ELISA and computational analysis RESULTS: The study identified 1467 proteins in the blood, of which nine were upregulated and 35 were downregulated by more than 2-fold. T-ALL patients showed a significant increase in specific disease-related proteins, such as eleven-nineteen lysine-rich leukemia protein, triggering receptor expressed on myeloid cells 1, cisplatin resistance-associated-overexpressed protein, X-ray radiation resistance-associated protein 1, tumor necrosis factor receptor superfamily member 10D, protein S100-A8, and copine-4, by more than 3-fold CONCLUSION: The findings of this study provide a valuable protein map of leukemic cells and identify potential biomarkers for leukemic aggressiveness. However, further studies using larger T-ALL patient samples must confirm these preliminary results.
Collapse
Affiliation(s)
- Vivek Singh
- King George's Medical University, Lucknow, UP, India
| | - Ranjana Singh
- King George's Medical University, Lucknow, UP, India,.
| | | |
Collapse
|
14
|
Yang X, Yang L, Luo A, Liu S, Zhang X, Liu X, Liu X, Luo A, Cai M, Yan Y, Wu X, Huang K, Xu L, Jiang H. IKZF3 polymorphisms contribute to the increased risk of acute lymphoblastic leukemia in children. Cancer 2024; 130:973-984. [PMID: 38018448 DOI: 10.1002/cncr.35129] [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: 08/18/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Acute lymphoblastic leukemia (ALL) is the most common cancer in children. IKZF3 (IKAROS family zinc finger 3) is a hematopoietic-specific transcription factor, and it has been validated that it is involved in leukemia. However, the role of IKZF3 single-nucleotide polymorphisms (SNPs) remains unclear. In this case-control study, the authors investigated the association of IKZF3 SNPs with ALL in children. METHODS Six IKZF3 reference SNPs (rs9635726, rs2060941, rs907092, rs12946510, rs1453559, and rs62066988) were genotyped in 692 patients who had ALL (cases) and in 926 controls. The associations between IKZF3 polymorphisms and ALL risk were determined using odds ratios (ORs) and 95% confidence intervals (CIs). The associations of rs9635726 and rs2060941 with the risk of ALL were further estimated by using false-positive report probability (FPRP) analysis. Functional analysis in silico was performed to evaluate the probability that rs9635726 and rs2060941 might influence the regulation of IKZF3. RESULTS The authors observed that rs9635726C>T (adjusted OR, 1.49; 95% CI, 1.06-2.11; p = .023) and rs2060941G>T (adjusted OR, 1.51; 95% CI, 1.24-1.84; p = .001) were related to and increased risk of ALL in the recessive and dominant models, respectively. Furthermore, the associations of both rs9635726 (FPRP = .177) and rs2060941 (FPRP < .001) with ALL were noteworthy in the FPRP analysis. Functional analysis indicated that rs9635726 and rs2060941 might repress the transcription of IKZF3 by disrupting its binding to MLLT1, TAF1, POLR2A, and/or RAD21. CONCLUSIONS This study revealed that IKZF3 polymorphisms were associated with increased ALL susceptibility in children and might influence the expression of IKZF3 by disrupting its binding to MLLT1, TAF1, POLR2A, and/or RAD21. IKZF3 polymorphisms were suggested as a biomarker for childhood ALL.
Collapse
Affiliation(s)
- Xu Yang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Lihua Yang
- Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Annie Luo
- Victoria College, University of Toronto, Toronto, Ontario, Canada
| | - Shanshan Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xiaohong Zhang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoping Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xiaodan Liu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Ailing Luo
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Mansi Cai
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Yaping Yan
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke Huang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ling Xu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Hua Jiang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
15
|
Weng N, Zhou C, Zhou Y, Zhong Y, Jia Z, Rao X, Qiu H, Zeng G, Jin X, Zhang J, Zhuang Z, Liang Z, Deng Y, Li Q, Yang S, Luo H, Wang H, Wu X. IKZF4/NONO-RAB11FIP3 axis promotes immune evasion in gastric cancer via facilitating PD-L1 endosome recycling. Cancer Lett 2024; 584:216618. [PMID: 38211652 DOI: 10.1016/j.canlet.2024.216618] [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: 07/04/2023] [Revised: 12/05/2023] [Accepted: 12/16/2023] [Indexed: 01/13/2024]
Abstract
As an immune checkpoint protein expressed by diverse cancer cells, programmed death ligand 1 (PD-L1) facilitates immune evasion by interacting with programmed cell death-1 (PD-1) on T cells. Despite the clinical benefits observed in various cancer types, strategies targeting PD-1/PD-L1 have demonstrated limited efficacy in gastric cancer (GC). Furthermore, the regulation of PD-L1, especially at post-translational modification levels, remains largely unknown. Therefore, it is crucial to elucidate the mechanisms governing PD-L1 expression to enhance anti-tumor immunity. In this study, we have identified that IKAROS family zinc finger 4 (IKZF4) and Non-POU domain-containing octamer-binding (NONO) synergistically regulate and enhance the expression of RAB11 family-interacting protein 3 (RAB11FIP3) in GC. The IKZF4/NONO-RAB11FIP3 axis facilitates the endosomal recycling of PD-L1, particularly on the cell membrane of GC cells. Moreover, overexpression of RAB11FIP3 mitigates the hypo-expression of PD-L1 protein resulting from IKZF4 or NONO deletion. Functionally, the silencing of RAB11FIP3 or IKZF4 promotes T cell proliferation, and enhances T-cell cytotoxicity towards GC cells in vitro, which further inhibits tumor immune evasion in mice via increasing the infiltration of CD8+ T cells into the tumor microenvironment (TME) to suppress GC progression. Our study suggests that the IKZF4/NONO-RAB11FIP3 axis promotes immune evasion by facilitating PD-L1 endosome recycling, thus presenting a potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Nuoqing Weng
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Chuzhou Zhou
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yuhang Zhou
- Department of Gastroenterology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yanping Zhong
- Department of Health Management, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Zhe Jia
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Xionghui Rao
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Huaiyu Qiu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Guangyan Zeng
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Xinghan Jin
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Jianbao Zhang
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Zhehong Zhuang
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Zhihao Liang
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yuan Deng
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Qinghai Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Shasha Yang
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Huixing Luo
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Huiyun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Xiaobin Wu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
16
|
Wu B, Ye N, Zhao K, Shi M, Liao J, Zhang J, Chen W, Li X, Han Y, Cortes-Clerget M, Regnier ML, Parmentier M, Mathes C, Rampf F, Gallou F. Implementation of micelle-enabled C(sp 2)-C(sp 3) cross-electrophile coupling in pharmaceutical synthesis. Chem Commun (Camb) 2024; 60:2349-2352. [PMID: 38284323 DOI: 10.1039/d3cc05916b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
A sustainable C(sp2)-C(sp3) cross-electrophile coupling was developed between readily available 5-bromophthalide and 1-benzyl-4-iodopiperidine under micellar conditions, leading to a key intermediate of one of our development compounds. Copper was found to play a crucial role as a co-catalyst in this dual catalysis system. The chemistry and process were successfully demonstrated in a kilo scale to deliver sufficient drug substance to the clinical campaigns. This is the first reported scale-up of such a challenging cross-electrophilic coupling that uses an aqueous medium, and not undesirable reprotoxic polar aprotic solvents (e.g. DMF, DMAc, and NMP).
Collapse
Affiliation(s)
- Bin Wu
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Ning Ye
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Kangming Zhao
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Min Shi
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Jiayu Liao
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Jing Zhang
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Wei Chen
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Xianzhong Li
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | - Yufeng Han
- Chemical & Analytical Development, Suzhou Novartis Technical Development Co., Ltd, Changshu, Jiangsu 215537, China.
| | | | | | - Michael Parmentier
- Chemical & Analytical Development, Novartis Pharma AG, 4056 Basel, Switzerland.
| | - Christian Mathes
- Chemical & Analytical Development, Novartis Pharma AG, 4056 Basel, Switzerland.
| | - Florian Rampf
- Chemical & Analytical Development, Novartis Pharma AG, 4056 Basel, Switzerland.
| | - Fabrice Gallou
- Chemical & Analytical Development, Novartis Pharma AG, 4056 Basel, Switzerland.
| |
Collapse
|
17
|
Kastner P, Aukenova A, Chan S. Evolution of the Ikaros family transcription factors: From a deuterostome ancestor to humans. Biochem Biophys Res Commun 2024; 694:149399. [PMID: 38134477 DOI: 10.1016/j.bbrc.2023.149399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023]
Abstract
Ikaros family proteins (Ikaros, Helios, Aiolos, Eos) are zinc finger transcription factors essential for the development and function of the adaptive immune system. They also control developmental events in neurons and other cell types, suggesting that they possess crucial functions across disparate cell types. These functions are likely shared among the organisms in which these factors exist, and it is thus important to obtain a view of their distribution and conservation across organisms. How this family evolved remains poorly understood. Here we mined protein, mRNA and DNA databases to identify proteins with DNA-binding domains homologous to that of Ikaros. We show that Ikaros-related proteins exist in organisms from all four deuterostome phyla (chordates, echinoderms, hemichordates, xenacoelomorpha), but not in more distant groups. While most non-vertebrates have a single family member, this family grew to six members in the acoel worm Hofstenia miamia, three in jawless and four in jawed vertebrates. Most residues involved in DNA contact from zinc fingers 2 to 4 were identical across the Ikaros family, suggesting conserved mechanisms for target sequence recognition. Further, we identified a novel KRKxxxPxK/R motif that inhibits DNA binding in vitro which was conserved across the deuterostome phyla. We also identified a EψψxxxψM(D/E)QAIxxAIxYLGA(D/E)xL motif conserved among human Ikaros, Aiolos, Helios and subsets of chordate proteins, and motifs that are specific to subsets of vertebrate family members. Some of these motifs are targets of mutations in human patients. Finally we show that the atypical family member Pegasus emerged only in vertebrates, which is consistent with its function in bone. Our data provide a novel evolutionary perspective for Ikaros family proteins and suggest that they have conserved regulatory functions across deuterostomes.
Collapse
Affiliation(s)
- Philippe Kastner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), ILLKIRCH, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, ILLKIRCH, France; Centre National de la Recherche Scientifique (CNRS), UMR7104, ILLKIRCH, France; Université de Strasbourg, ILLKIRCH, France; Faculté de Médecine, Université de Strasbourg, Strasbourg, France.
| | - Adina Aukenova
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), ILLKIRCH, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, ILLKIRCH, France; Centre National de la Recherche Scientifique (CNRS), UMR7104, ILLKIRCH, France; Université de Strasbourg, ILLKIRCH, France
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), ILLKIRCH, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, ILLKIRCH, France; Centre National de la Recherche Scientifique (CNRS), UMR7104, ILLKIRCH, France; Université de Strasbourg, ILLKIRCH, France.
| |
Collapse
|
18
|
Nishiguchi G, Mascibroda LG, Young SM, Caine EA, Abdelhamed S, Kooijman JJ, Miller DJ, Das S, McGowan K, Mayasundari A, Shi Z, Barajas JM, Hiltenbrand R, Aggarwal A, Chang Y, Mishra V, Narina S, Thomas M, Loughran AJ, Kalathur R, Yu K, Zhou S, Wang X, High AA, Peng J, Pruett-Miller SM, Daniels DL, Urh M, Shelat AA, Mullighan CG, Riching KM, Zaman GJR, Fischer M, Klco JM, Rankovic Z. Selective CK1α degraders exert antiproliferative activity against a broad range of human cancer cell lines. Nat Commun 2024; 15:482. [PMID: 38228616 PMCID: PMC10791743 DOI: 10.1038/s41467-024-44698-1] [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: 05/01/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Molecular-glue degraders are small molecules that induce a specific interaction between an E3 ligase and a target protein, resulting in the target proteolysis. The discovery of molecular glue degraders currently relies mostly on screening approaches. Here, we describe screening of a library of cereblon (CRBN) ligands against a panel of patient-derived cancer cell lines, leading to the discovery of SJ7095, a potent degrader of CK1α, IKZF1 and IKZF3 proteins. Through a structure-informed exploration of structure activity relationship (SAR) around this small molecule we develop SJ3149, a selective and potent degrader of CK1α protein in vitro and in vivo. The structure of SJ3149 co-crystalized in complex with CK1α + CRBN + DDB1 provides a rationale for the improved degradation properties of this compound. In a panel of 115 cancer cell lines SJ3149 displays a broad antiproliferative activity profile, which shows statistically significant correlation with MDM2 inhibitor Nutlin-3a. These findings suggest potential utility of selective CK1α degraders for treatment of hematological cancers and solid tumors.
Collapse
Affiliation(s)
- Gisele Nishiguchi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Lauren G Mascibroda
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sarah M Young
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Elizabeth A Caine
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Sherif Abdelhamed
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | | | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sourav Das
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kevin McGowan
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anand Mayasundari
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zhe Shi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Juan M Barajas
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ryan Hiltenbrand
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anup Aggarwal
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yunchao Chang
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Vibhor Mishra
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Shilpa Narina
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Melvin Thomas
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Allister J Loughran
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ravi Kalathur
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Suiping Zhou
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Memphis, TN, 38105, USA
| | - Danette L Daniels
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Marjeta Urh
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kristin M Riching
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Guido J R Zaman
- Oncolines B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Marcus Fischer
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
19
|
da Silva Lima F, da Silva Gonçalves CE, Fock RA. A review of the role of zinc finger proteins on hematopoiesis. J Trace Elem Med Biol 2023; 80:127290. [PMID: 37659124 DOI: 10.1016/j.jtemb.2023.127290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/04/2023]
Abstract
The bone marrow is responsible for producing an incredible number of cells daily in order to maintain blood homeostasis through a process called hematopoiesis. Hematopoiesis is a greatly demanding process and one entirely dependent on complex interactions between the hematopoietic stem cell (HSC) and its surrounding microenvironment. Zinc (Zn2+) is considered an important trace element, playing diverse roles in different tissues and cell types, and zinc finger proteins (ZNF) are proteins that use Zn2+ as a structural cofactor. In this way, the ZNF structure is supported by a Zn2+ that coordinates many possible combinations of cysteine and histidine, with the most common ZNF being of the Cys2His2 (C2H2) type, which forms a family of transcriptional activators that play an important role in different cellular processes such as development, differentiation, and suppression, all of these being essential processes for an adequate hematopoiesis. This review aims to shed light on the relationship between ZNF and the regulation of the hematopoietic tissue. We include works with different designs, including both in vitro and in vivo studies, detailing how ZNF might regulate hematopoiesis.
Collapse
Affiliation(s)
- Fabiana da Silva Lima
- Department of Food and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
20
|
Eckardt JN, Stasik S, Röllig C, Petzold A, Sauer T, Scholl S, Hochhaus A, Crysandt M, Brümmendorf TH, Naumann R, Steffen B, Kunzmann V, Einsele H, Schaich M, Burchert A, Neubauer A, Schäfer-Eckart K, Schliemann C, Krause SW, Herbst R, Hänel M, Hanoun M, Kaiser U, Kaufmann M, Rácil Z, Mayer J, Oelschlägel U, Berdel WE, Ehninger G, Serve H, Müller-Tidow C, Platzbecker U, Baldus CD, Dahl A, Schetelig J, Bornhäuser M, Middeke JM, Thiede C. Mutated IKZF1 is an independent marker of adverse risk in acute myeloid leukemia. Leukemia 2023; 37:2395-2403. [PMID: 37833543 PMCID: PMC10681898 DOI: 10.1038/s41375-023-02061-1] [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: 05/28/2023] [Revised: 09/24/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023]
Abstract
Genetic lesions of IKZF1 are frequent events and well-established markers of adverse risk in acute lymphoblastic leukemia. However, their function in the pathophysiology and impact on patient outcome in acute myeloid leukemia (AML) remains elusive. In a multicenter cohort of 1606 newly diagnosed and intensively treated adult AML patients, we found IKZF1 alterations in 45 cases with a mutational hotspot at N159S. AML with mutated IKZF1 was associated with alterations in RUNX1, GATA2, KRAS, KIT, SF3B1, and ETV6, while alterations of NPM1, TET2, FLT3-ITD, and normal karyotypes were less frequent. The clinical phenotype of IKZF1-mutated AML was dominated by anemia and thrombocytopenia. In both univariable and multivariable analyses adjusting for age, de novo and secondary AML, and ELN2022 risk categories, we found mutated IKZF1 to be an independent marker of adverse risk regarding complete remission rate, event-free, relapse-free, and overall survival. The deleterious effects of mutated IKZF1 also prevailed in patients who underwent allogeneic hematopoietic stem cell transplantation (n = 519) in both univariable and multivariable models. These dismal outcomes are only partially explained by the hotspot mutation N159S. Our findings suggest a role for IKZF1 mutation status in AML risk modeling.
Collapse
Affiliation(s)
- Jan-Niklas Eckardt
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany.
| | - Sebastian Stasik
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Christoph Röllig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Andreas Petzold
- Dresden-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Tim Sauer
- German Cancer Research Center (DKFZ) and Medical Clinic V, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Scholl
- Klinik für Innere Medizin II, Jena University Hospital, Jena, Germany
| | - Andreas Hochhaus
- Klinik für Innere Medizin II, Jena University Hospital, Jena, Germany
| | - Martina Crysandt
- Department of Hematology, Oncology, Hemostaseology, and Cell Therapy, University Hospital RWTH Aachen, Aachen, Germany
| | - Tim H Brümmendorf
- Department of Hematology, Oncology, Hemostaseology, and Cell Therapy, University Hospital RWTH Aachen, Aachen, Germany
| | - Ralph Naumann
- Medical Clinic III, St. Marien-Hospital Siegen, Siegen, Germany
| | - Björn Steffen
- Medical Clinic II, University Hospital Frankfurt, Frankfurt (Main), Germany
| | - Volker Kunzmann
- Medical Clinic and Policlinic II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Medical Clinic and Policlinic II, University Hospital Würzburg, Würzburg, Germany
| | - Markus Schaich
- Department of Hematology, Oncology and Palliative Care, Rems-Murr-Hospital Winnenden, Winnenden, Germany
| | - Andreas Burchert
- Department of Hematology, Oncology and Immunology, Philipps-University-Marburg, Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, Philipps-University-Marburg, Marburg, Germany
| | - Kerstin Schäfer-Eckart
- Department of Internal Medicine V, Paracelsus Medizinische Privatuniversität and University Hospital Nuremberg, Nuremberg, Germany
| | | | - Stefan W Krause
- Medical Clinic V, University Hospital Erlangen, Erlangen, Germany
| | - Regina Herbst
- Medical Clinic III, Chemnitz Hospital AG, Chemnitz, Germany
| | - Mathias Hänel
- Medical Clinic III, Chemnitz Hospital AG, Chemnitz, Germany
| | - Maher Hanoun
- Department of Hematology, University Hospital Essen, Essen, Germany
| | - Ulrich Kaiser
- Medical Clinic II, St. Bernward Hospital, Hildesheim, Germany
| | - Martin Kaufmann
- Department of Hematology, Oncology and Palliative Care, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Zdenek Rácil
- Department of Internal Medicine, Hematology and Oncology, Masaryk University Hospital, Brno, Czech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, Masaryk University Hospital, Brno, Czech Republic
| | - Uta Oelschlägel
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Wolfgang E Berdel
- Department of Medicine A, University Hospital Münster, Münster, Germany
| | - Gerhard Ehninger
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Hubert Serve
- Medical Clinic II, University Hospital Frankfurt, Frankfurt (Main), Germany
| | - Carsten Müller-Tidow
- German Cancer Research Center (DKFZ) and Medical Clinic V, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Platzbecker
- Medical Clinic I Hematology and Celltherapy, University Hospital Leipzig, Leipzig, Germany
| | - Claudia D Baldus
- Department of Internal Medicine, University Hospital Kiel, Kiel, Germany
| | - Andreas Dahl
- Dresden-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Johannes Schetelig
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- DKMS Clinical Trials Unit, Dresden, Germany
| | - Martin Bornhäuser
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
- German Consortium for Translational Cancer Research DKFZ, Heidelberg, Germany
- National Center for Tumor Disease (NCT), Dresden, Germany
| | - Jan Moritz Middeke
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Christian Thiede
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
21
|
Mohajeri A, Vaseghi-Shanjani M, Rosenfeld JA, Yang GX, Lu H, Sharma M, Lin S, Salman A, Waqas M, Sababi Azamian M, Worley KC, Del Bel KL, Kozak FK, Rahmanian R, Biggs CM, Hildebrand KJ, Lalani SR, Nicholas SK, Scott DA, Mostafavi S, van Karnebeek C, Henkelman E, Halparin J, Yang CL, Armstrong L, Turvey SE, Lehman A. Dominant negative variants in IKZF2 cause ICHAD syndrome, a new disorder characterised by immunodysregulation, craniofacial anomalies, hearing impairment, athelia and developmental delay. J Med Genet 2023; 60:1092-1104. [PMID: 37316189 PMCID: PMC11206234 DOI: 10.1136/jmg-2022-109127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/29/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Helios (encoded by IKZF2), a member of the Ikaros family of transcription factors, is a zinc finger protein involved in embryogenesis and immune function. Although predominantly recognised for its role in the development and function of T lymphocytes, particularly the CD4+ regulatory T cells (Tregs), the expression and function of Helios extends beyond the immune system. During embryogenesis, Helios is expressed in a wide range of tissues, making genetic variants that disrupt the function of Helios strong candidates for causing widespread immune-related and developmental abnormalities in humans. METHODS We performed detailed phenotypic, genomic and functional investigations on two unrelated individuals with a phenotype of immune dysregulation combined with syndromic features including craniofacial differences, sensorineural hearing loss and congenital abnormalities. RESULTS Genome sequencing revealed de novo heterozygous variants that alter the critical DNA-binding zinc fingers (ZFs) of Helios. Proband 1 had a tandem duplication of ZFs 2 and 3 in the DNA-binding domain of Helios (p.Gly136_Ser191dup) and Proband 2 had a missense variant impacting one of the key residues for specific base recognition and DNA interaction in ZF2 of Helios (p.Gly153Arg). Functional studies confirmed that both these variant proteins are expressed and that they interfere with the ability of the wild-type Helios protein to perform its canonical function-repressing IL2 transcription activity-in a dominant negative manner. CONCLUSION This study is the first to describe dominant negative IKZF2 variants. These variants cause a novel genetic syndrome characterised by immunodysregulation, craniofacial anomalies, hearing impairment, athelia and developmental delay.
Collapse
Affiliation(s)
- Arezoo Mohajeri
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Maryam Vaseghi-Shanjani
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Gui Xiang Yang
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Henry Lu
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Mehul Sharma
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Susan Lin
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Areesha Salman
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Meriam Waqas
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Mahshid Sababi Azamian
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Kim C Worley
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Kate L Del Bel
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Frederick K Kozak
- Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ronak Rahmanian
- Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine M Biggs
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Kyla J Hildebrand
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Seema R Lalani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Sarah K Nicholas
- Department of Pediatrics, Texas Children's Hospital, Houston, Texas, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Sara Mostafavi
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Clara van Karnebeek
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Erika Henkelman
- Department of Surgery, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Jessica Halparin
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Connie L Yang
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Linlea Armstrong
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
- Provincial Medical Genetics Program, BC Children's & Women's Hosp, Vancouver, British Columbia, Canada
| | - Stuart E Turvey
- Department of Pediatrics, The University of British Columbia and BC Children's Hospital, Vancouver, British Columbia, Canada
| | - Anna Lehman
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Kimura H, Onozawa M, Yoshida S, Miyashita N, Yokoyama S, Matsukawa T, Hirabayashi S, Goto H, Endo T, Oguri S, Fujisawa S, Mori A, Kondo T, Hidaka D, Okada K, Ota S, Kakinoki Y, Tsutsumi Y, Yamamoto S, Miyagishima T, Hashiguchi J, Nagashima T, Ibata M, Wakasa K, Haseyama Y, Fujimoto K, Ishihara T, Sakai H, Teshima T. Dominant-negative type of IKZF1 deletion showed a favorable prognosis in adult B-cell acute lymphoblastic leukemia. Ann Hematol 2023; 102:3103-3113. [PMID: 37597110 DOI: 10.1007/s00277-023-05405-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
IKZF1 deletion is a recurrent genomic alteration in B-cell acute lymphoblastic leukemia (B-ALL) and is divided into dominant-negative (DN) and loss of function (LOF) deletions. The prognostic impact of each deletion has not been fully elucidated. We retrospectively analyzed 117 patients with adult B-ALL including 60 patients with BCR::ABL1-positive B-ALL and 57 patients with BCR::ABL1-negative B-ALL by the fluorescence in situ hybridization (FISH) method for IKZF1 deletion and multiplex PCR for the 4 most common IKZF1 deletions (∆4-7, ∆2-7, ∆2-8, and ∆4-8). Samples, in which IKZF1 deletion was detected by FISH but a specific type of deletion was not identified by the PCR, were categorized as "other." Patients were classified into a DN group that had at least 1 allele of ∆4-7 (n = 23), LOF and other group (n = 40), and wildtype group (n = 54). DN type IKZF1 deletions were found in 33.3% of BCR::ABL1-positive cases and 5.2% of BCR::ABL1-negative cases. LOF and other type IKZF1 deletions were found in 43.4% of BCR::ABL1-positive cases and 24.6% of BCR::ABL1-negative cases. Patients with the DN group showed significantly higher overall survival (OS) than that of the LOF and other and WT groups (P = 0.011). Multivariate analysis including age, WBC counts, complex karyotype, and DN type IKZF1 deletion showed that the DN type of IKZF1 deletion (HR = 0.22, P = 0.013) had a positive impact and age ≥ 65 (HR = 1.92, P = 0.029) had a negative impact on OS. The prognostic impact of IKZF1 deletion depends on the type of deletion and DN type of IKZF1 deletion showed better prognosis in adult B-ALL patients.Clinical trial registration This study was part of a prospective observational study (Hokkaido Leukemia Net, UMIN000048611). It was conducted in compliance with ethical principles based on the Helsinki Declaration and was approved by the institutional review board of Hokkaido University Hospital (#015-0344).
Collapse
Affiliation(s)
- Hiroyuki Kimura
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Masahiro Onozawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan.
| | - Shota Yoshida
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Naoki Miyashita
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Shota Yokoyama
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Toshihiro Matsukawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | | | - Hideki Goto
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Tomoyuki Endo
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
| | - Satoshi Oguri
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Shinichi Fujisawa
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Akio Mori
- Blood Disorders Center, Aiiku Hospital, Sapporo, Japan
| | - Takeshi Kondo
- Blood Disorders Center, Aiiku Hospital, Sapporo, Japan
| | - Daisuke Hidaka
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Kohei Okada
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | | | - Yutaka Tsutsumi
- Department of Hematology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Satoshi Yamamoto
- Department of Hematology, Sapporo City General Hospital, Sapporo, Japan
| | | | - Junichi Hashiguchi
- Department of Internal Medicine, Kitami Red Cross Hospital, Kitami, Japan
| | - Takahiro Nagashima
- Department of Internal Medicine, Kitami Red Cross Hospital, Kitami, Japan
| | - Makoto Ibata
- Department of Hematology, Sapporo Kosei General Hospital, Sapporo, Japan
| | - Kentaro Wakasa
- Department of Hematology, Obihiro Kosei Hospital, Obihiro, Japan
| | | | - Katsuya Fujimoto
- Department of Hematology, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | | | - Hajime Sakai
- Department of Hematology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Graduate School of Medicine, Kita 15, Nishi 7, Kita-Ku, Sapporo, 0608638, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
23
|
Richardson PG, Trudel S, Popat R, Mateos MV, Vangsted AJ, Ramasamy K, Martinez-Lopez J, Quach H, Orlowski RZ, Arnao M, Lonial S, Karanes C, Pawlyn C, Kim K, Oriol A, Berdeja JG, Rodríguez Otero P, Casas-Avilés I, Spirli A, Poon J, Li S, Gong J, Wong L, Lamba M, Pierce DW, Amatangelo M, Peluso T, Maciag P, Katz J, Pourdehnad M, Bahlis NJ. Mezigdomide plus Dexamethasone in Relapsed and Refractory Multiple Myeloma. N Engl J Med 2023; 389:1009-1022. [PMID: 37646702 DOI: 10.1056/nejmoa2303194] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
BACKGROUND Despite recent progress, multiple myeloma remains incurable. Mezigdomide is a novel cereblon E3 ubiquitin ligase modulator with potent antiproliferative and tumoricidal activity in preclinical models of multiple myeloma, including those resistant to lenalidomide and pomalidomide. METHODS In this phase 1-2 study, we administered oral mezigdomide in combination with dexamethasone to patients with relapsed and refractory myeloma. The primary objectives of phase 1 (dose-escalation cohort) were to assess safety and pharmacokinetics and to identify the dose and schedule for phase 2. In phase 2 (dose-expansion cohort), objectives included the assessment of the overall response (partial response or better), safety, and efficacy of mezigdomide plus dexamethasone at the dose and schedule determined in phase 1. RESULTS In phase 1, a total of 77 patients were enrolled in the study. The most common dose-limiting toxic effects were neutropenia and febrile neutropenia. On the basis of the phase 1 findings, investigators determined the recommended phase 2 dose of mezigdomide to be 1.0 mg, given once daily in combination with dexamethasone for 21 days, followed by 7 days off, in each 28-day cycle. In phase 2, a total of 101 patients received the dose identified in phase 1 in the same schedule. All patients in the dose-expansion cohort had triple-class-refractory multiple myeloma, 30 patients (30%) had received previous anti-B-cell maturation antigen (anti-BCMA) therapy, and 40 (40%) had plasmacytomas. The most common adverse events, almost all of which proved to be reversible, included neutropenia (in 77% of the patients) and infection (in 65%; grade 3, 29%; grade 4, 6%). No unexpected toxic effects were encountered. An overall response occurred in 41% of the patients (95% confidence interval [CI], 31 to 51), the median duration of response was 7.6 months (95% CI, 5.4 to 9.5; data not mature), and the median progression-free survival was 4.4 months (95% CI, 3.0 to 5.5), with a median follow-up of 7.5 months (range, 0.5 to 21.9). CONCLUSIONS The all-oral combination of mezigdomide plus dexamethasone showed promising efficacy in patients with heavily pretreated multiple myeloma, with treatment-related adverse events consisting mainly of myelotoxic effects. (Funded by Celgene, a Bristol-Myers Squibb Company; CC-92480-MM-001 ClinicalTrials.gov number, NCT03374085; EudraCT number, 2017-001236-19.).
Collapse
Affiliation(s)
- Paul G Richardson
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Suzanne Trudel
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Rakesh Popat
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - María-Victoria Mateos
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Annette J Vangsted
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Karthik Ramasamy
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Joaquín Martinez-Lopez
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Hang Quach
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Robert Z Orlowski
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Mario Arnao
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Sagar Lonial
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Chatchada Karanes
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Charlotte Pawlyn
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Kihyun Kim
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Albert Oriol
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Jesus G Berdeja
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Paula Rodríguez Otero
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Ignacio Casas-Avilés
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Alessia Spirli
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Jennifer Poon
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Shaoyi Li
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Jing Gong
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Lilly Wong
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Manisha Lamba
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Daniel W Pierce
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Michael Amatangelo
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Teresa Peluso
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Paulo Maciag
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Jessica Katz
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Michael Pourdehnad
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| | - Nizar J Bahlis
- From Dana-Farber Cancer Institute, Boston (P.G.R.); the Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto (S.T.), and Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB (N.J.B.) - both in Canada; NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust (R.P.), the Institute of Cancer Research (C.P.), and the Royal Marsden NHS Foundation Trust (C.P.), London, and the Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford (K.R.) - all in the United Kingdom; University Hospital of Salamanca/IBSAL, Salamanca (M.-V.M.), the Department of Hematology, Hospital 12 de Octubre, Department of Medicine, School of Medicine, Complutense University, H12O-CNIO Clinical Research Unit, CIBERONC, Madrid (J.M.-L.), Hospital Universitari La Fe, Valencia (M.A.), Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Badalona (A.O.), Clínica Universidad de Navarra, CIMA, IDISNA, CIBERONC, Pamplona (P.R.O.), and Hospital San Pedro de Alcántara, Cáceres (I.C.-A.) - all in Spain; the Department of Hematology, Rigshospitalet, Copenhagen (A.J.V.); St. Vincent's Hospital Melbourne, University of Melbourne, Melbourne, VIC, Australia (H.Q.); the Department of Lymphoma and Myeloma, University of Texas M.D. Anderson Cancer Center, Houston (R.Z.O.); Winship Cancer Institute, Emory University, Atlanta (S. Lonial); Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA (C.K.); Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea (K.K.); Sarah Cannon Research Institute, Nashville (J.G.B.); Celgene International, a Bristol-Myers Squibb Company, Boudry, Switzerland (A.S., T.P.); and Bristol Myers Squibb, Princeton, NJ (J.P., S. Li, J.G., L.W., M.L., D.W.P., M.A., P.M., J.K., M.P.)
| |
Collapse
|
24
|
Chang J, Yamashita M, Padhi AK, Zhang KYJ, Taniuchi I. Impaired tissue homing by the Ikzf3 N159S variant is mediated by interfering with Ikaros function. Front Immunol 2023; 14:1239779. [PMID: 37662955 PMCID: PMC10469740 DOI: 10.3389/fimmu.2023.1239779] [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: 06/14/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
AIOLOS, encoded by IKZF3, is a member of the IKZF family of proteins that plays an important role in regulating late B-cell differentiation. Human individuals heterozygous for the AIOLOS p.N160S variant displayed impaired humoral immune responses as well as impaired B and T cell development. We have previously reported that a mouse strain harboring an Ikzf3N159S allele that corresponds to human IKZF3N160S recapitulated immune-deficient phenotypes, such as impaired B cell development and loss of CD23 expression. In this study, we investigated the effect of the Ikzf3N159S variant and found that B1a cell development was impaired in Ikzf3N159S/N159S mice. In addition, CD62L expression was severely decreased in both B and T lymphocytes by the Ikzf3N159S mutation, in a dose-dependent manner. Mixed bone marrow chimera experiments have revealed that most immunodeficient phenotypes, including low CD62L expression, occur in intrinsic cells. Interestingly, while Ikzf3N159S/N159S lymphocytes were still present in the spleen, they were completely outcompeted by control cells in the lymph nodes, suggesting that the capacity for homing or retention in the lymph nodes was lost due to the Ikzf3N159S mutation. The homing assay confirmed severely decreased homing abilities to lymph nodes of Ikzf3N159S/N159S B and T lymphocytes but selective enrichment of CD62L expressing Ikzf3N159S/N159S lymphocytes in lymph nodes. This finding suggests that impaired CD62L expression is the major reason for the impaired homing capacity caused by the Ikzf3N159S mutation. Interestingly, an excess amount of Ikaros, but not Aiolos, restored CD62L expression in Ikzf3N159S/N159S B cells. Together with the loss of CD62L expression due to Ikaros deficiency, the AiolosN159S mutant protein likely interferes with Ikaros function through heterodimerization, at least in activating the Sell gene encoding CD62L expression. Thus, our results revealed that AiolosN159S causes some immunodeficient phenotypes via the pathogenesis referred to as the heterodimeric interference as observed for AiolosG158R variant.
Collapse
Affiliation(s)
- Jingjie Chang
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Motoi Yamashita
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Aditya K. Padhi
- Laboratory for Structural Bioinformatics, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Kam Y. J. Zhang
- Laboratory for Structural Bioinformatics, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| |
Collapse
|
25
|
Tuazon JA, Read KA, Sreekumar BK, Roettger JE, Yaeger MJ, Varikuti S, Pokhrel S, Jones DM, Warren RT, Powell MD, Rasheed MN, Duncan EG, Childs LM, Gowdy KM, Oestreich KJ. Eos Promotes TH2 Differentiation by Interacting with and Propagating the Activity of STAT5. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:365-376. [PMID: 37314436 PMCID: PMC10524986 DOI: 10.4049/jimmunol.2200861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/18/2023] [Indexed: 06/15/2023]
Abstract
The Ikaros zinc-finger transcription factor Eos has largely been associated with sustaining the immunosuppressive functions of regulatory T cells. Paradoxically, Eos has more recently been implicated in promoting proinflammatory responses in the dysregulated setting of autoimmunity. However, the precise role of Eos in regulating the differentiation and function of effector CD4+ T cell subsets remains unclear. In this study, we find that Eos is a positive regulator of the differentiation of murine CD4+ TH2 cells, an effector population that has been implicated in both immunity against helminthic parasites and the induction of allergic asthma. Using murine in vitro TH2 polarization and an in vivo house dust mite asthma model, we find that EosKO T cells exhibit reduced expression of key TH2 transcription factors, effector cytokines, and cytokine receptors. Mechanistically, we find that the IL-2/STAT5 axis and its downstream TH2 gene targets are one of the most significantly downregulated pathways in Eos-deficient cells. Consistent with these observations, we find that Eos forms, to our knowledge, a novel complex with and supports the tyrosine phosphorylation of STAT5. Collectively, these data define a regulatory mechanism whereby Eos propagates STAT5 activity to facilitate TH2 cell differentiation.
Collapse
Affiliation(s)
- Jasmine A. Tuazon
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, 43210; USA
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, OH, 43210; USA
| | - Kaitlin A. Read
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, 43210; USA
| | | | - Jack E. Roettger
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, 43210; USA
| | - Michael J. Yaeger
- Division of Pulmonary, Critical Care and Sleep Medicine; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
| | - Sanjay Varikuti
- Division of Pulmonary, Critical Care and Sleep Medicine; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
| | - Srijana Pokhrel
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
| | - Devin M. Jones
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, 43210; USA
| | - Robert T. Warren
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
| | - Michael D. Powell
- Department of Microbiology and Immunology; Emory University School of Medicine, Atlanta, GA, 30322; USA
| | - Mustafa N. Rasheed
- Department of Emergency Medicine; Emory University Medical Center, Atlanta, GA, 30322; USA
| | | | - Lauren M. Childs
- Department of Mathematics; Virginia Tech, Blacksburg, VA, 24061; USA
| | - Kymberly M. Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
| | - Kenneth J. Oestreich
- Department of Microbial Infection and Immunity; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
- Pelotonia Institute for Immuno-Oncology; The Ohio State Comprehensive Cancer Center, Columbus, Ohio, 43210; USA
- Infectious Diseases Institute; The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, 43210; USA
| |
Collapse
|
26
|
Palanichamy JK, Tran TM, King JK, Katzman S, Ritter AJ, Sharma G, Tso C, Contreras JR, Fernando TR, Sanford JR, Rao DS. Distinct oncogenic phenotypes in hematopoietic specific deletions of Trp53. Sci Rep 2023; 13:7490. [PMID: 37160922 PMCID: PMC10169790 DOI: 10.1038/s41598-023-33949-8] [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: 08/27/2022] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
Loss of function in the tumor suppressor gene TP53 is the most common alteration seen in human cancer. In mice, P53 deletion in all cells leads predominantly to the development of T-cell lymphomas, followed by B-cell lymphomas, sarcomas and teratomas. In order to dissect the role of P53 in the hematopoietic system, we generated and analyzed two different mouse models deficient for P53. A pan-hematopoietic P53 deletion mouse was created using Vav1-Cre based deletion; and a B-cell-specific deletion mouse was created using a CD19-Cre based deletion. The Vav1-P53CKO mice predominantly developed T-cell malignancies in younger mice, and myeloid malignancies in older mice. In T-cell malignancies, there was accelerated thymic cell maturation with overexpression of Notch1 and its downstream effectors. CD19-P53CKO mice developed marginal zone expansion in the spleen, followed by marginal zone lymphoma, some of which progressed to diffuse large B-cell lymphomas. Interestingly, marginal zone and diffuse large B-cell lymphomas had a unique gene expression signature characterized by activation of the PI3K pathway, compared with wild type marginal zone or follicular cells of the spleen. This study demonstrates lineage specific P53 deletion leading to distinct phenotypes secondary to unique gene expression programs set in motion.
Collapse
Affiliation(s)
| | - Tiffany M Tran
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
- Molecular, Cellular and Integrative Physiology Graduate Program, UCLA, Los Angeles, USA
| | | | - Sol Katzman
- Center for Biomolecular Science & Engineering, UCSC, Santa Cruz, USA
| | - Alexander J Ritter
- Department of Molecular, Cell and Developmental Biology and Center for Molecular Biology of RNA, UCSC, Santa Cruz, USA
| | - Gunjan Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Christine Tso
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Jorge R Contreras
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | | | - Jeremy R Sanford
- Department of Molecular, Cell and Developmental Biology and Center for Molecular Biology of RNA, UCSC, Santa Cruz, USA
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, USA.
- Broad Stem Cell Research Center, UCLA, Los Angeles, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 650 Charles E Young Drive, Los Angeles, CA, 90095, USA.
| |
Collapse
|
27
|
Wang Y, Zhao H, He Y, Zhang P, Zeng C, Du T, Shen Q, Chen Y, Zhao S. IKZF4 acts as a novel tumor suppressor in non-small cell lung cancer by suppressing Notch signaling pathway. Cell Signal 2023; 107:110679. [PMID: 37044192 DOI: 10.1016/j.cellsig.2023.110679] [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: 02/07/2023] [Revised: 03/24/2023] [Accepted: 04/08/2023] [Indexed: 04/14/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the predominant cause of cancer-related mortality globally, although many clinical efforts have been developed to improve the outcomes. The Ikaros zing-finger family transcription factors (IKZFs) have been proved to play pivotal roles in lymphopoiesis and myeloma progression, but their roles in solid tumors development remain unclear. We performed integrative bioinformatical analysis to determine the dysregulation expression of IKZFs in multiple tumors and the correlation between IKZF4 and NSCLC tumor environment. We showed that IKZFs were dysregulated in multiple tumors and IKZF4 was significantly decreased in NSCLC tissues and cell lines due to promoter hypermethylation. We found that low IKZF4 expression obviously correlated with patients' poor clinical outcome. We revealed that IKZF4 overexpression inhibited NSCLC cell growth, migration and xenograft tumor growth, supporting the inhibitory role of IKZF4 in NSCLC tumorigenesis. Additionally, integrative bioinformatical analysis showed that IKZF4 was involved in NSCLC tumor microenvironment. Mechanically, RNA-seq results showed that IKZF4 forced-expression remarkably suppressed Notch signaling pathway in NSCLC, which was validated by qRT-PCR and immunoblot assays. Moreover, we screened several potential agonists for IKZF4.
Collapse
Affiliation(s)
- Yanbo Wang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Hanqing Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yaomei He
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Yunnan Key Laboratory of Natural Medicinal Chemistry, Kunming 650201, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou 450052, China
| | - Cheng Zeng
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tongxuan Du
- Institute of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Qiushuo Shen
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Institute of Biomedical Engineering, Kunming Medical University, Kunming, Yunnan 650500, China.
| | - Yongbin Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
28
|
Mok CC. Targeted Small Molecules for Systemic Lupus Erythematosus: Drugs in the Pipeline. Drugs 2023; 83:479-496. [PMID: 36972009 PMCID: PMC10042116 DOI: 10.1007/s40265-023-01856-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Despite the uncertainty of the pathogenesis of systemic lupus erythematosus, novel small molecules targeting specific intracellular mechanisms of immune cells are being developed to reverse the pathophysiological processes. These targeted molecules have the advantages of convenient administration, lower production costs, and the lack of immunogenicity. The Janus kinases, Bruton's tyrosine kinases, and spleen tyrosine kinases are important enzymes for activating downstream signals from various receptors on immune cells that include cytokines, growth factor, hormones, Fc, CD40, and B-cell receptors. Suppression of these kinases impairs cellular activation, differentiation, and survival, leading to diminished cytokine actions and autoantibody secretion. Intracellular protein degradation by immunoproteasomes, levered by the cereblon E3 ubiquitin ligase complex, is an essential process for the regulation of cellular functions and survival. Modulation of the immunoproteasomes and cereblon leads to depletion of long-lived plasma cells, reduced plasmablast differentiation, and production of autoantibodies and interferon-α. The sphingosine 1-phosphate/sphingosine 1-phosphate receptor-1 pathway is responsible for lymphocyte trafficking, regulatory T-cell/Th17 cell homeostasis, and vascular permeability. Sphingosine 1-phosphate receptor-1 modulators limit the trafficking of autoreactive lymphocytes across the blood-brain barrier, increase regulatory T-cell function, and decrease production of autoantibodies and type I interferons. This article summarizes the development of these targeted small molecules in the treatment of systemic lupus erythematosus, and the future prospect for precision medicine.
Collapse
Affiliation(s)
- Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Tsing Chung Koon Road, New Territories, Hong Kong SAR, China.
| |
Collapse
|
29
|
Li H, Ye M, Hu Z, Lu H, Zheng D, Wu M, Ge T, Xu S, Ge Z, Zhang S, Xu G, Chen H. IKZF3 is a novel prognostic biomarker for head and neck squamous cell carcinoma: A study based on bioinformatics analysis. Medicine (Baltimore) 2023; 102:e33124. [PMID: 36930079 PMCID: PMC10019242 DOI: 10.1097/md.0000000000033124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/08/2023] [Indexed: 03/18/2023] Open
Abstract
In the past few years, immunotherapy of tumors has become an extensive research hotspot, and the value of IKZF family genes in the tumor microenvironment has also been increasingly recognized. However, the expression of the IKAROS family zinc finger 3 (IKZF3) gene in human head and neck squamous cell carcinoma (HNSCC) and its prognostic value were not reported for the main subset until now. In the present study, we analyzed the relationship between IKZF3 gene expression and the survival of HNSCC patients. To evaluate the potential of IKZF3 as a prognostic biomarker for HNSCC comprehensively, multiple online analysis tools, including UALCAN, cBioPortal, GEPIA, WebGestalt, String, Genomic Data Commons, and TIMER databases were utilized in our study. We observed that the HNSCC patients with higher IKZF3 expression tended to exhibit longer overall survival. Univariate and multivariate Cox regression analyses indicated that age and grade were independent prognostic indicators in HNSCC. Moreover, Gene Ontology and KEGG function enrichment analyses showed that several pathways in HNSCC might be pivotal pathways regulated by IKZF3, which revealed that IKZF3 was probably participating in the occurrence and development of HNSCC. Furthermore, the hypomethylation of the IKZF3 gene was closely associated with genes that observed mutation in HNSCC. IKZF3 was significantly correlated with several immune cells in HNSCC (e.g., CD8+ T cell, CD4+ cell, and dendritic cell). We explored the potential prognostic values and roles of the IKZF3 in HNSCC, revealing that IKZF3 was probably a novel and reliable prognostic biomarker for patients with HNSCC.
Collapse
Affiliation(s)
- Hongxiang Li
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Mengmeng Ye
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Zeyang Hu
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Haoxuan Lu
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Dawei Zheng
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Mi Wu
- Department of Emergency, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Ting Ge
- Department of Respiratory, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Shuguang Xu
- Department of Respiratory, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Zhen Ge
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China
| | - Shuoni Zhang
- Department of Emergency, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Guodong Xu
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Hang Chen
- Department of Cardiothoracic Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
30
|
Bonazzi S, d'Hennezel E, Beckwith REJ, Xu L, Fazal A, Magracheva A, Ramesh R, Cernijenko A, Antonakos B, Bhang HEC, Caro RG, Cobb JS, Ornelas E, Ma X, Wartchow CA, Clifton MC, Forseth RR, Fortnam BH, Lu H, Csibi A, Tullai J, Carbonneau S, Thomsen NM, Larrow J, Chie-Leon B, Hainzl D, Gu Y, Lu D, Meyer MJ, Alexander D, Kinyamu-Akunda J, Sabatos-Peyton CA, Dales NA, Zécri FJ, Jain RK, Shulok J, Wang YK, Briner K, Porter JA, Tallarico JA, Engelman JA, Dranoff G, Bradner JE, Visser M, Solomon JM. Discovery and characterization of a selective IKZF2 glue degrader for cancer immunotherapy. Cell Chem Biol 2023; 30:235-247.e12. [PMID: 36863346 DOI: 10.1016/j.chembiol.2023.02.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 12/15/2022] [Accepted: 02/09/2023] [Indexed: 03/04/2023]
Abstract
Malignant tumors can evade destruction by the immune system by attracting immune-suppressive regulatory T cells (Treg) cells. The IKZF2 (Helios) transcription factor plays a crucial role in maintaining function and stability of Treg cells, and IKZF2 deficiency reduces tumor growth in mice. Here we report the discovery of NVP-DKY709, a selective molecular glue degrader of IKZF2 that spares IKZF1/3. We describe the recruitment-guided medicinal chemistry campaign leading to NVP-DKY709 that redirected the degradation selectivity of cereblon (CRBN) binders from IKZF1 toward IKZF2. Selectivity of NVP-DKY709 for IKZF2 was rationalized by analyzing the DDB1:CRBN:NVP-DKY709:IKZF2(ZF2 or ZF2-3) ternary complex X-ray structures. Exposure to NVP-DKY709 reduced the suppressive activity of human Treg cells and rescued cytokine production in exhausted T-effector cells. In vivo, treatment with NVP-DKY709 delayed tumor growth in mice with a humanized immune system and enhanced immunization responses in cynomolgus monkeys. NVP-DKY709 is being investigated in the clinic as an immune-enhancing agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Simone Bonazzi
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA.
| | - Eva d'Hennezel
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA.
| | | | - Lei Xu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Aleem Fazal
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Anna Magracheva
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Radha Ramesh
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | - Hyo-Eun C Bhang
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Jennifer S Cobb
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Xiaolei Ma
- Novartis Institutes for Biomedical Research, Emeryville, CA, USA
| | | | | | - Ry R Forseth
- Novartis Institutes for Biomedical Research, East Hanover, NJ, USA
| | | | - Hongbo Lu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Alfredo Csibi
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Jennifer Tullai
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Seth Carbonneau
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Noel M Thomsen
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Jay Larrow
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Dominik Hainzl
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Yi Gu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Darlene Lu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Matthew J Meyer
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Dylan Alexander
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | - Natalie A Dales
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Rishi K Jain
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Janine Shulok
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Y Karen Wang
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Karin Briner
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | | | - Glenn Dranoff
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - James E Bradner
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Michael Visser
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | |
Collapse
|
31
|
Yan J, Zheng Z. Discovery of Highly Potent CRBN Ligands and Insight into Their Binding Mode through Molecular Docking and Molecular Dynamics Simulations. ChemMedChem 2023; 18:e202200573. [PMID: 36750890 DOI: 10.1002/cmdc.202200573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/21/2022] [Indexed: 02/09/2023]
Abstract
Cereblon (CRBN) is a substrate receptor of E3 ubiquitin ligase as well as the target of thalidomide and lenalidomide, plays a vital role in endogenous protein degradation. In this article, two series of compounds with novel structure were designed, synthesized and evaluated against CRBN. YJ1b, designed based on our previous finding, shown strong binding affinity toward CRBN (IC50 =0.206 μM) by forming a salt bridge interaction with amino acid residue Glu377 of CRBN, it was 13-fold compared with that of lenalidomide (IC50 =2.694 μM) in TR-FRET assay. YJ2c and YJ2h, two analogs of YJ1b, also exhibit high binding affinity toward CRBN (IC50 =0.211 μM and IC50 =0.282 μM, respectively). While, molecular docking and 100 ns molecular dynamic simulation studies were conducted to insight into the unique binding mode of YJ1b, YJ2c and YJ2e toward CRBN. The new compounds with special binding mode in this article may serve for the further optimization and discovery of novel high potent CRBN ligands.
Collapse
Affiliation(s)
- Jian Yan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Zhibing Zheng
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| |
Collapse
|
32
|
Conserva MR, Redavid I, Anelli L, Zagaria A, Tarantini F, Cumbo C, Tota G, Parciante E, Coccaro N, Minervini CF, Minervini A, Specchia G, Musto P, Albano F. IKAROS in Acute Leukemia: A Positive Influencer or a Mean Hater? Int J Mol Sci 2023; 24:3282. [PMID: 36834692 PMCID: PMC9961161 DOI: 10.3390/ijms24043282] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
One key process that controls leukemogenesis is the regulation of oncogenic gene expression by transcription factors acting as tumor suppressors. Understanding this intricate mechanism is crucial to elucidating leukemia pathophysiology and discovering new targeted treatments. In this review, we make a brief overview of the physiological role of IKAROS and the molecular pathway that contributes to acute leukemia pathogenesis through IKZF1 gene lesions. IKAROS is a zinc finger transcription factor of the Krüppel family that acts as the main character during hematopoiesis and leukemogenesis. It can activate or repress tumor suppressors or oncogenes, regulating the survival and proliferation of leukemic cells. More than 70% of Ph+ and Ph-like cases of acute lymphoblastic leukemia exhibit IKZF1 gene variants, which are linked to worse treatment outcomes in both childhood and adult B-cell precursor acute lymphoblastic leukemia. In the last few years, much evidence supporting IKAROS involvement in myeloid differentiation has been reported, suggesting that loss of IKZF1 might also be a determinant of oncogenesis in acute myeloid leukemia. Considering the complicated "social" network that IKAROS manages in hematopoietic cells, we aim to focus on its involvement and the numerous alterations of molecular pathways it can support in acute leukemias.
Collapse
Affiliation(s)
- Maria Rosa Conserva
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Immacolata Redavid
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Luisa Anelli
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Antonella Zagaria
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Francesco Tarantini
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Cosimo Cumbo
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Giuseppina Tota
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Elisa Parciante
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Nicoletta Coccaro
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Crescenzio Francesco Minervini
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Angela Minervini
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Giorgina Specchia
- School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Pellegrino Musto
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| | - Francesco Albano
- Hematology Section, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari ‘Aldo Moro’, 70124 Bari, Italy
| |
Collapse
|
33
|
Protein Kinase CK2 and Epstein-Barr Virus. Biomedicines 2023; 11:biomedicines11020358. [PMID: 36830895 PMCID: PMC9953236 DOI: 10.3390/biomedicines11020358] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Protein kinase CK2 is a pleiotropic protein kinase, which phosphorylates a number of cellular and viral proteins. Thereby, this kinase is implicated in the regulation of cellular signaling, controlling of cell proliferation, apoptosis, angiogenesis, immune response, migration and invasion. In general, viruses use host signaling mechanisms for the replication of their genome as well as for cell transformation leading to cancer. Therefore, it is not surprising that CK2 also plays a role in controlling viral infection and the generation of cancer cells. Epstein-Barr virus (EBV) lytically infects epithelial cells of the oropharynx and B cells. These latently infected B cells subsequently become resting memory B cells when passing the germinal center. Importantly, EBV is responsible for the generation of tumors such as Burkitt's lymphoma. EBV was one of the first human viruses, which was connected to CK2 in the early nineties of the last century. The present review shows that protein kinase CK2 phosphorylates EBV encoded proteins as well as cellular proteins, which are implicated in the lytic and persistent infection and in EBV-induced neoplastic transformation. EBV-encoded and CK2-phosphorylated proteins together with CK2-phosphorylated cellular signaling proteins have the potential to provide efficient virus replication and cell transformation. Since there are powerful inhibitors known for CK2 kinase activity, CK2 might become an attractive target for the inhibition of EBV replication and cell transformation.
Collapse
|
34
|
Meng C, Chen S, He Q, Tan J, Wu J, Zhao J. IKZF3 modulates cerebral ischemia/reperfusion injury by inhibiting neuroinflammation. Int Immunopharmacol 2023; 114:109480. [PMID: 36525791 DOI: 10.1016/j.intimp.2022.109480] [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: 09/28/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Neuroinflammation is a key mediator to the pathogenic cascades induced by cerebral ischemia-reperfusion (I/R) injury. IKZF3, a key zinc finger transcription factor in the Ikaros family, has already been shown to modulate a wide range of cell functions and the production of inflammatory mediators. However, the effects of IKZF3 on inflammation and the potential mechanism after cerebral I/R injury remain unclear. In this study, we evaluated the effect of IKZF3 on HT-22 cells under oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro and in mice with MACO in vivo. We found that IKZF3 expression peaked at 12 h after MCAO and OGD/R, and there was high expression of IKZF3 in brain tissues and HT-22 cells. IKZF3 knockdown exacerbated the damage by OGD-induced HT-22 cells injury and MCAO-induced brain injury in mice by regulating the production of inflammatory factors, which promoted the phosphorylation and nuclear transfer of NF-ĸB and may bind with NF-ĸB-p65 in vivo and in vitro. Our results suggested that IKZF3 may provide a new target in improve neurological recovery and reducing neuroinflammation after cerebral I/R injury.
Collapse
Affiliation(s)
- Changchang Meng
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Shiyu Chen
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China
| | - Qi He
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Junyi Tan
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Jingxian Wu
- Department of Pathology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| | - Jing Zhao
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China; Institute of Neuroscience, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
35
|
Chen LY, Gooding S. Tumor and microenvironmental mechanisms of resistance to immunomodulatory drugs in multiple myeloma. Front Oncol 2022; 12:1038329. [PMID: 36439455 PMCID: PMC9682014 DOI: 10.3389/fonc.2022.1038329] [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: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 10/07/2023] Open
Abstract
Resistance to immunomodulatory drugs (IMiDs®) is a major cause of treatment failure, disease relapse and ultimately poorer outcomes in multiple myeloma (MM). In order to optimally deploy IMiDs and their newer derivates CRBN E3 ligase modulators (CELMoDs®) into future myeloma therapeutic regimens, it is imperative to understand the mechanisms behind the inevitable emergence of IMiD resistance. IMiDs bind and modulate Cereblon (CRBN), the substrate receptor of the CUL4CRBN E3 ubiquitin ligase, to target novel substrate proteins for ubiquitination and degradation. Most important of these are IKZF1 and IKZF3, key MM survival transcription factors which sustain the expression of myeloma oncogenes IRF4 and MYC. IMiDs directly target MM cell proliferation, but also stimulate T/NK cell activation by their CRBN-mediated effects, and therefore enhance anti-MM immunity. Thus, their benefits in myeloma are directed against tumor and immune microenvironment - and in considering the mechanisms by which IMiD resistance emerges, both these effects must be appraised. CRBN-dependent mechanisms of IMiD resistance, including CRBN genetic aberrations, CRBN protein loss and CRBN-substrate binding defects, are beginning to be understood. However, only a proportion of IMiD-resistant cases are related to CRBN and therefore additional mechanisms, which are currently less well described, need to be sought. These include resistance within the immune microenvironment. Here we review the existing evidence on both tumor and immune microenvironment mechanisms of resistance to IMiDs, pose important questions for future study, and consider how knowledge regarding resistance mechanism may be utilized to guide treatment decision making in the clinic.
Collapse
Affiliation(s)
- Lucia Y. Chen
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
| | - Sarah Gooding
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
- Oxford Centre for Translational Myeloma Research, University of Oxford, Oxford, United Kingdom
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
36
|
Yang LK, Lin CX, Li SH, Liang JJ, Xiao LL, Xie GH, Liu HW, Liao X. Novel IKZF3 transcriptomic signature correlates with positive outcomes of skin cutaneous melanoma: A pan-cancer analysis. Front Genet 2022; 13:1036402. [PMID: 36353107 PMCID: PMC9638148 DOI: 10.3389/fgene.2022.1036402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
To investigate the potential relationship between Ikaros family genes and skin cutaneous melanoma (SKCM), we undertook a pan-cancer analysis of the transcriptional signature and clinical data of melanoma through multiple databases. First, 10,327 transcriptomic samples from different cancers were included to determine the overall characteristics and clinical prognoses associated with Ikaros gene expression across cancer types. Second, differentially expressed genes analysis, prognostic evaluation, and gene set enrichment analysis were employed to investigate the role of Ikaros (IKZF) genes in SKCM. Third, we evaluated the relationship between Ikaros family genes and SKCM immune infiltrates and verified the findings using the GEO single-cell sequencing dataset. The results show that Ikaros genes were widely expressed among different cancer types with independently similar patterns as follows: 1. IKZF1 and IKZF3, and 2. IKZF2 and IKZF4–5. IKZF2 and IKZF5 were downregulated in the primary tumor, and IKZF1–3 expression decreased significantly as the T-stage or metastasis increased in SKCM. Moreover, high IKZF1–3 expression was associated with better overall survival, disease-specific survival, and progression-free interval. IKZF3 is an independent prognostic factor of SKCM. Among Ikaros genes, the expression of IKZF1 and IKZF3 positively correlated with the infiltration level of CD4+ T cells and CD8+ T cells, B cells, and Tregs in SKCM and negatively correlated with the infiltration level of M0 and M1 macrophages. Moreover, single-cell sequencing data analysis revealed that IKZF1 and IKZF3 were mainly expressed by immune cells. Correlation analysis shows the immune factors and drug responses associated with IKZF3 expression. In conclusion, the present study is the first, to our knowledge, to identify a pan-cancer genomic signature of the Ikaros gene family among different cancers. Expression of these family members, particularly high levels of IKZF3, indicate positive immunological status and beneficial clinical outcomes of SKCM. IKZF3 may therefore serve as potential targets for immunotherapy of melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xuan Liao
- *Correspondence: Hong-Wei Liu, ; Xuan Liao,
| |
Collapse
|
37
|
Lipsky PE, Vollenhoven RV, Dörner T, Werth VP, Merrill JT, Furie R, Petronijevic M, Velasco Zamora B, Majdan M, Irazoque-Palazuelos F, Terbrueggen R, Delev N, Weiswasser M, Korish S, Stern M, Hersey S, Ye Y, Gaudy A, Liu Z, Gagnon R, Tang S, Schafer PH. Biological impact of iberdomide in patients with active systemic lupus erythematosus. Ann Rheum Dis 2022; 81:annrheumdis-2022-222212. [PMID: 35477518 PMCID: PMC9279852 DOI: 10.1136/annrheumdis-2022-222212] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/10/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Iberdomide is a high-affinity cereblon ligand that promotes proteasomal degradation of transcription factors Ikaros (IKZF1) and Aiolos (IKZF3). Pharmacodynamics and pharmacokinetics of oral iberdomide were evaluated in a phase 2b study of patients with active systemic lupus erythematosus (SLE). METHODS Adults with autoantibody-positive SLE were randomised to placebo (n=83) or once daily iberdomide 0.15 mg (n=42), 0.3 mg (n=82) or 0.45 mg (n=81). Pharmacodynamic changes in whole blood leucocytes were measured by flow cytometry, regulatory T cells (Tregs) by epigenetic assay, plasma cytokines by ultrasensitive cytokine assay and gene expression by Modular Immune Profiling. RESULTS Iberdomide exhibited linear pharmacokinetics and dose-dependently modulated leucocytes and cytokines. Compared with placebo at week 24, iberdomide 0.45 mg significantly (p<0.001) reduced B cells, including those expressing CD268 (TNFRSF13C) (-58.3%), and plasmacytoid dendritic cells (-73.9%), and increased Tregs (+104.9%) and interleukin 2 (IL-2) (+144.1%). Clinical efficacy was previously reported in patients with high IKZF3 expression and high type I interferon (IFN) signature at baseline and confirmed here in those with an especially high IFN signature. Iberdomide decreased the type I IFN gene signature only in patients with high expression at baseline (-81.5%; p<0.001) but decreased other gene signatures in all patients. CONCLUSION Iberdomide significantly reduced activity of type I IFN and B cell pathways, and increased IL-2 and Tregs, suggesting a selective rebalancing of immune abnormalities in SLE. Clinical efficacy corresponded to reduction of the type I IFN gene signature. TRIAL REGISTRATION NUMBER NCT03161483.
Collapse
Affiliation(s)
- Peter E Lipsky
- RILITE Foundation and AMPEL BioSolutions, Charlottesville, Virginia, USA
| | | | - Thomas Dörner
- German Rheumatism Research Center, Charité University Hospital, Berlin, Germany
| | - Victoria P Werth
- University of Pennsylvania and the Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Joan T Merrill
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Richard Furie
- Department of Rheumatology, Northwell Health, Great Neck, New York, USA
| | | | | | - Maria Majdan
- Samodzielny Publiczny Szpital Kliniczny Nr 4 w Lublinie, Medical University of Lublin, Lublin, Poland
| | | | | | | | | | | | - Mark Stern
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Sarah Hersey
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Ying Ye
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Zhaohui Liu
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Shaojun Tang
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | | |
Collapse
|
38
|
Yamashita M, Morio T. AIOLOS Variants Causing Immunodeficiency in Human and Mice. Front Immunol 2022; 13:866582. [PMID: 35444653 PMCID: PMC9014263 DOI: 10.3389/fimmu.2022.866582] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
AIOLOS is encoded by IKZF3 and is a member of the IKAROS zinc finger transcription factor family. Heterozygous missense variants in the second zinc finger of AIOLOS have recently been reported to be found in the families of patients with inborn errors of immunity. The AIOLOSG159R variant was identified in patients with B-lymphopenia and familial Epstein–Barr virus-associated lymphoma. Early B-cell progenitors were significantly reduced in the bone marrow of patients with AIOLOSG159R. Another variant, AIOLOSN160S was identified in the patients presented with hypogammaglobulinemia, susceptibility to Pneumocystis jirovecii pneumonia, and chronic lymphocytic leukemia. Patients with AIOLOSN160S had mostly normal B cell counts but showed increased levels of CD21lo B cells, decreased CD23 expression, and abrogated CD40 response. Both variants were determined to be loss-of-function. Mouse models harboring the corresponding patient’s variants recapitulated the phenotypes of the patients. AIOLOS is therefore a novel disease-causing gene in human adaptive immune deficiency.
Collapse
Affiliation(s)
- Motoi Yamashita
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
39
|
Richardson PG, Mateos MV, Vangsted AJ, Ramasamy K, Abildgaard N, Ho PJ, Quach H, Bahlis NJ. The role of E3 ubiquitin ligase in multiple myeloma: potential for cereblon E3 ligase modulators in the treatment of relapsed/refractory disease. Expert Rev Proteomics 2022; 19:235-246. [PMID: 36342226 DOI: 10.1080/14789450.2022.2142564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Insights into the mechanisms of protein homeostasis and proteasomal degradation have led to new strategies of redirecting the ubiquitin-proteasome system (UPS) to reduce or eliminate proteins or survival factors key to malignant pathobiology, multiple myeloma (MM) in particular. These strategies have enabled researchers to target proteins that were previously considered difficult to modulate by pharmacological means. AREAS COVERED This review provides a brief overview of UPS biology, particularly the role of the CRL4CRBN E3 ubiquitin ligase complex, and summarizes current strategies for co-opting the UPS, including CELMoD compounds, SNIPERs, PROTACs, and degronimids. A detailed discussion is provided on lead CELMoD compounds iberdomide and mezigdomide, which are currently being evaluated in clinical trials in patients with MM. EXPERT OPINION Since a high proportion of patients develop drug resistance, it is vital to have novel therapeutic agents for treating relapsed patients with MM more effectively. It is encouraging that the expanding pathophysiological insight into cellular signaling pathways in MM increasingly translates into the development of novel therapeutic agents such as targeted protein degraders. This holds promise for improving outcomes in MM and beyond.
Collapse
Affiliation(s)
- Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Karthik Ramasamy
- Department of Clinical Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Niels Abildgaard
- Hematology Research Unit, Department of Hematology, Odense University Hospital, Odense, Denmark; and Department of Clinical Research.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - P Joy Ho
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Hang Quach
- Department of Haematology, St Vincent's Hospital, Melbourne, Australia
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Merrill JT, Werth VP, Furie R, van Vollenhoven R, Dörner T, Petronijevic M, Velasco J, Majdan M, Irazoque-Palazuelos F, Weiswasser M, Korish S, Ye Y, Gaudy A, Schafer PH, Liu Z, Agafonova N, Delev N. Phase 2 Trial of Iberdomide in Systemic Lupus Erythematosus. N Engl J Med 2022; 386:1034-1045. [PMID: 35294813 DOI: 10.1056/nejmoa2106535] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Iberdomide, a cereblon modulator promoting degradation of the transcription factors Ikaros and Aiolos, which affect leukocyte development and autoimmunity, is being evaluated for the treatment of systemic lupus erythematosus (SLE). METHODS In this phase 2 trial, we randomly assigned patients in a 2:2:1:2 ratio to receive oral iberdomide (at a dose of 0.45, 0.30, or 0.15 mg) or placebo once daily for 24 weeks, in addition to standard medications. The primary end point at week 24 was a response on the SLE Responder Index (SRI-4), which was defined as a reduction of at least 4 points in the Systemic Lupus Erythematosus Disease Activity Index 2000 score (a 24-item weighted score of lupus activity that ranges from 0 to 105, with higher scores indicating greater disease activity), no new disease activity as measured on the British Isles Lupus Assessment Group 2004 index, and no increase of 0.3 points or more in the Physician's Global Assessment score (on a visual-analogue scale ranging from 0 [no disease activity] to 3 [maximal disease]). RESULTS A total of 288 patients received the assigned intervention: 81 received iberdomide at a dose of 0.45 mg, 82 received iberdomide at a dose of 0.30 mg, 42 received iberdomide at a dose of 0.15 mg, and 83 received placebo. At week 24, the percentages of patients with an SRI-4 response were 54% in the iberdomide 0.45-mg group, 40% in the iberdomide 0.30-mg group, 48% in the iberdomide 0.15-mg group, and 35% in the placebo group (adjusted difference between the iberdomide 0.45-mg group and the placebo group, 19.4 percentage points; 95% confidence interval, 4.1 to 33.4; P = 0.01), with no significant differences between the groups that received the lower doses of iberdomide and the group that received placebo. Iberdomide-associated adverse events included urinary tract and upper respiratory tract infections and neutropenia. CONCLUSIONS In this 24-week, phase 2 trial involving patients with SLE, iberdomide at a dose of 0.45 mg resulted in a higher percentage of patients with an SRI-4 response than did placebo. Data from larger, longer trials are needed to determine the efficacy and safety of iberdomide in SLE. (Funded by Bristol Myers Squibb; ClinicalTrials.gov number, NCT03161483; EudraCT number, 2016-004574-17.).
Collapse
Affiliation(s)
- Joan T Merrill
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Victoria P Werth
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Richard Furie
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Ronald van Vollenhoven
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Thomas Dörner
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Milan Petronijevic
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Jorge Velasco
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Maria Majdan
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Fedra Irazoque-Palazuelos
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Michael Weiswasser
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Shimon Korish
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Ying Ye
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Allison Gaudy
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Peter H Schafer
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Zhaohui Liu
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Nataliya Agafonova
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| | - Nikolay Delev
- From the Oklahoma Medical Research Foundation, Oklahoma City (J.T.M.); the University of Pennsylvania and the Corporal Michael J. Crescenz VA Medical Center - both in Philadelphia (V.P.W.); Northwell Health, Great Neck, NY (R.F.); Amsterdam University Medical Centers, Amsterdam (R.V.); Charité-Universitätsmedizin, Berlin (T.D.); the Military Medical Academy, Belgrade, Serbia (M.P.); Instituto Centro de Enfermedades Reumáticas, Buenos Aires (J.V.); Independent Public Clinical Hospital Number 4, Medical University of Lublin, Lublin, Poland (M.M.); Centro de Investigación y Tratamiento Reumatológico, Mexico City, Mexico (F.I.-P.); and Bristol Myers Squibb, Princeton, NJ (M.W., S.K., Y.Y., A.G., P.H.S., Z.L., N.A., N.D.)
| |
Collapse
|
41
|
Furie RA, Hough DR, Gaudy A, Ye Y, Korish S, Delev N, Weiswasser M, Zhan X, Schafer PH, Werth VP. Iberdomide in patients with systemic lupus erythematosus: a randomised, double-blind, placebo-controlled, ascending-dose, phase 2a study. Lupus Sci Med 2022; 9:9/1/e000581. [PMID: 35169036 PMCID: PMC8852715 DOI: 10.1136/lupus-2021-000581] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/14/2022] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To evaluate safety, pharmacokinetics, pharmacodynamics and efficacy of iberdomide in patients with SLE. Iberdomide is a high-affinity cereblon ligand that targets the hematopoietic transcription factors Ikaros and Aiolos for proteasomal degradation. METHODS A 12-week, multicentre, double-blind, placebo-controlled, dose-escalation study in active SLE was followed by a 2-year, open-label active treatment extension phase (ATEP) (NCT02185040). In the dose-escalation phase, adults with active SLE were randomised to oral placebo or iberdomide (0.3 mg every other day, 0.3 mg once daily, 0.6 mg and 0.3 mg alternating once daily, or 0.6 mg once daily). Primary endpoints were safety and tolerability. RESULTS The dose-escalation phase enrolled 42 patients, with 33 completing this phase and 17 patients enrolling into the ATEP. In the dose-escalation phase, the most common treatment-emergent adverse events (TEAEs; iberdomide/placebo groups) were nausea (20.6%/12.5%), diarrhoea (17.6%/12.5%) and upper respiratory tract infection (11.8%/12.5%). Most TEAEs were mild or moderate in severity and more common in the highest dose groups in both study phases. In the dose-escalation phase, Physician's Global Assessment and Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI) activity scores improved relative to baseline and placebo in all iberdomide groups, with a trend toward continued score improvements in the ATEP. In the dose-escalation phase, iberdomide treatment resulted in dose-dependent reductions in total B cells and plasmacytoid dendritic cells in blood. Improvements in CLASI activity scores correlated with plasmacytoid dendritic cell depletion. CONCLUSIONS These proof-of-concept findings suggest a favourable benefit/risk ratio in SLE for iberdomide, a drug with a novel immunomodulatory mechanism of action, supporting further clinical investigation.
Collapse
Affiliation(s)
| | - Douglas R Hough
- Clinical Research, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Allison Gaudy
- Translational Development, Clinical Pharmacology, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Ying Ye
- ICF Early Clinical Development, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Shimon Korish
- Clinical R&D, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Nikolay Delev
- Clinical R&D, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Xiaojiang Zhan
- Biometrics, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Peter H Schafer
- TRC Inflammation, CV & Fibrosis and Global Health, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Victoria P Werth
- Corporal Michael J Crescenz VA Medical Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Zou Y, Liu B, Li L, Yin Q, Tang J, Jing Z, Huang X, Zhu X, Chi T. IKZF3 deficiency potentiates chimeric antigen receptor T cells targeting solid tumors. Cancer Lett 2022; 524:121-130. [PMID: 34687790 DOI: 10.1016/j.canlet.2021.10.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been successful in treating hematological malignancy, but solid tumors remain refractory. Here, we demonstrated that knocking out transcription factor IKZF3 in HER2-specific CAR T cells targeting breast cancer cells did not affect CAR expression or CAR T cell differentiation, but markedly enhanced killing of the cancer cells in vitro and in a xenograft model, which was associated with increased T cell activation and proliferation. Furthermore, IKZF3 KO had similar effects on the CD133-specific CAR T cells targeting glioblastoma cells. AlphaLISA and RNA-seq analyses indicate that IKZF3 KO increased the expression of genes involved in cytokine signaling, chemotaxis and cytotoxicity. Our results suggest a general strategy for enhancing CAR T efficacy on solid tumors.
Collapse
Affiliation(s)
- Yan Zou
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Bo Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Long Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Qinan Yin
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, Henan, 471000, China
| | - Jiaxing Tang
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Zhengyu Jing
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xuekai Zhu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China.
| | - Tian Chi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; Department of Immunobiology, Yale University Medical School, New Haven, CT, USA.
| |
Collapse
|
43
|
Ueta M. Pathogenesis of Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis With Severe Ocular Complications. Front Med (Lausanne) 2021; 8:651247. [PMID: 34869401 PMCID: PMC8635481 DOI: 10.3389/fmed.2021.651247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Stevens-Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN) is an acute inflammatory vesiculobullous reaction of the mucosa of the ocular surface, oral cavity, and genitals, and of the skin. Severe ocular complications (SOC) are observed in about half of SJS/TEN patients diagnosed by dermatologists and in burn units. Ophthalmologists treat SOC, and they tend to encounter the patients not only in the acute stage, but also in the chronic stage. Our investigation of the pathogenesis of SJS/TEN with SOC led us to suspect that abnormal innate mucosal immunity contributes to the ocular surface inflammation seen in SJS/TEN with SOC. We confirmed that cold medicines such as NSAIDs and multi-ingredient cold medications are the main causative drugs for SJS/TEN with SOC. Single nucleotide polymorphism (SNP) association analysis of cold medicine-related SJS/TEN with SOC showed that the Toll-like receptor 3 (TLR3)-, the prostaglandin-E receptor 3 (PTGER3)-, and the IKZF1 gene were significantly associated with SNPs and that these genes could regulate mucocutaneous inflammation including that of the ocular surface. We also examined the tear cytokines of SJS/TEN with SOC in the chronic stage and found that IL-8, IL-6, IFN-γ, RANTES, eotaxin, and MIP-1β were significantly upregulated in SJS/TEN with SOC in the chronic stage. Only IP-10 was significantly downregulated in SJS/TEN with SOC in the chronic stage. This mini-review summarizes the pathological mechanisms that we identified as underlying the development of SJS/TEN with SOC.
Collapse
Affiliation(s)
- Mayumi Ueta
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
44
|
Shahin T, Kuehn HS, Shoeb MR, Gawriyski L, Giuliani S, Repiscak P, Hoeger B, Yüce Petronczki Ö, Bal SK, Zoghi S, Dmytrus J, Seruggia D, Castanon I, Rezaei N, Varjosalo M, Halbritter F, Rosenzweig SD, Boztug K. Germline biallelic mutation affecting the transcription factor Helios causes pleiotropic defects of immunity. Sci Immunol 2021; 6:eabe3981. [PMID: 34826259 DOI: 10.1126/sciimmunol.abe3981] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Tala Shahin
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Hye Sun Kuehn
- Immunology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20814, USA
| | - Mohamed R Shoeb
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Lisa Gawriyski
- Institute of Biotechnology, Helsinki Institute of Life Science, Proteomics Unit, University of Helsinki, Helsinki, Finland
| | - Sarah Giuliani
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Peter Repiscak
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Birgit Hoeger
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Özlem Yüce Petronczki
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Sevgi Köstel Bal
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Samaneh Zoghi
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Jasmin Dmytrus
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Davide Seruggia
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Irinka Castanon
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science, Proteomics Unit, University of Helsinki, Helsinki, Finland
| | | | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20814, USA
| | - Kaan Boztug
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.,Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.,St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
Zaini A, Zaph C. Aiolos: a molecular guardian of type 2 innate immune cell residency and response. Mucosal Immunol 2021; 14:1221-1223. [PMID: 34408274 DOI: 10.1038/s41385-021-00444-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 02/04/2023]
Affiliation(s)
- Aidil Zaini
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Colby Zaph
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
46
|
Charliński G, Vesole DH, Jurczyszyn A. Rapid Progress in the Use of Immunomodulatory Drugs and Cereblon E3 Ligase Modulators in the Treatment of Multiple Myeloma. Cancers (Basel) 2021; 13:4666. [PMID: 34572892 PMCID: PMC8468542 DOI: 10.3390/cancers13184666] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the improvement in our understanding of the biology of MM and the introduction of new drug classes, including immunomodulatory drugs (IMiDs), proteasome inhibitors (PI), and monoclonal antibodies (MoAb), have significantly improved outcomes. The first IMiD introduced to treat MM was thalidomide. The side effects observed during treatment with thalidomide initiated work on the synthesis of IMiD analogs. Subsequently, lenalidomide and pomalidomide were developed, both with different safety profiles, and they have better tolerability than thalidomide. In 2010, the cereblon (CRBN) protein was discovered as a direct target of IMiDs. By binding to CRBN, IMiDs change the substrate specificity of the CRBN E3 ubiquitin ligase complex, which results in the breakdown of internal Ikaros and Aiolos proteins. Most clinical trials conducted, both in newly diagnosed, post-transplant maintenance and relapsed/refractory MM, report a beneficial effect of IMiDs on the extension of progression-free survival and overall survival in patients with MM. Due to side effects, thalidomide is used less frequently. Currently, lenalidomide is used at every phase of MM treatment. Lenalidomide is used in conjunction with other agents such as PIs and MoAb as induction and relapsed therapy. Pomalidomide is currently used to treat relapsed/refractory MM, also with PIs and monoclonal antibodies. Current clinical trials are evaluating the efficacy of IMiD derivatives, the CRBN E3 ligase modulators (CELMoDs). This review focuses on the impact of IMiDs for the treatment of MM.
Collapse
Affiliation(s)
- Grzegorz Charliński
- Department of Hematology, Warmian-Masurian Cancer Center of The Ministry of The Interior and Administration’s Hospital, 10-228 Olsztyn, Poland;
| | - David H. Vesole
- John Theurer Cancer Center at Hackensack Meridian School of Medicine, Hackensack, NJ 07601, USA;
| | - Artur Jurczyszyn
- Plasma Cell Dyscrasia Center, Department of Hematology, Faculty of Medicine, Jagiellonian University Medical College, 31-501 Kraków, Poland
| |
Collapse
|
47
|
de Melo TRF, Dulmovits BM, Fernandes GFDS, de Souza CM, Lanaro C, He M, Al Abed Y, Chung MC, Blanc L, Costa FF, Dos Santos JL. Synthesis and pharmacological evaluation of pomalidomide derivatives useful for sickle cell disease treatment. Bioorg Chem 2021; 114:105077. [PMID: 34130111 PMCID: PMC8387409 DOI: 10.1016/j.bioorg.2021.105077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Fetal hemoglobin (HbF) induction constitutes a valuable and validated approach to treat the symptoms of sickle cell disease (SCD). Here, we synthesized pomalidomide-nitric oxide (NO) donor derivatives (3a-f) and evaluated their suitability as novel HbF inducers. All compounds demonstrated different capacities of releasing NO, ranging 0.3-30.3%. Compound 3d was the most effective HbF inducer for CD34+ cells, exhibiting an effect similar to that of hydroxyurea. We investigated the mode of action of compound 3d for HbF induction by studying the in vitro alterations in the levels of transcription factors (BCL11A, IKAROS, and LRF), inhibition of histone deacetylase enzymes (HDAC-1 and HDAC-2), and measurement of cGMP levels. Additionally, compound 3d exhibited a potent anti-inflammatory effect similar to that of pomalidomide by reducing the TNF-α levels in human mononuclear cells treated with lipopolysaccharides up to 58.6%. Chemical hydrolysis studies revealed that compound 3d was stable at pH 7.4 up to 24 h. These results suggest that compound 3d is a novel HbF inducer prototype with the potential to treat SCD symptoms.
Collapse
Affiliation(s)
| | - Brian M Dulmovits
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | | | - Cristiane M de Souza
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Carolina Lanaro
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Minghzu He
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Yousef Al Abed
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA; Laboratory of Developmental Erythropoiesis, Les Nelkin Memorial Pediatric Oncology Laboratory, The Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
| | - Man Chin Chung
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil
| | - Lionel Blanc
- Department of Molecular Medicine and Pediatrics, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Fernando Ferreira Costa
- Faculty of Medical Sciences, State University of Campinas - UNICAMP, Campinas 13083-970, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara 14800-903, Brazil.
| |
Collapse
|
48
|
Xie S, Wei H, Peng A, Xie A, Li J, Fang C, Shi F, Yang Q, Huang H, Xie H, Pan X, Tian X, Huang J. Ikzf2 Regulates the Development of ICOS + Th Cells to Mediate Immune Response in the Spleen of S. japonicum-Infected C57BL/6 Mice. Front Immunol 2021; 12:687919. [PMID: 34475870 PMCID: PMC8406689 DOI: 10.3389/fimmu.2021.687919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background Th cells (helper T cells) have multiple functions in Schistosoma japonicum (S. japonicum) infection. Inducible co-stimulator (ICOS) is induced and expressed in activated T lymphocytes, which enhances the development of B cells and antibody production through the ICOS/ICOSL pathway. It remains unclear about the role and possible regulating mechanism of ICOS+ Th cells in the spleen of S. japonicum-infected C57BL/6 mice. Methods C57BL/6 mice were infected with cercariae of S. japonicum through the abdomen. The expression of ICOS, activation markers, and the cytokine production on CD4+ ICOS+ Th cells were detected by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Moreover, the differentially expressed gene data of ICOS+ and ICOS- Th cells from the spleen of infected mice were obtained by mRNA sequencing. Besides, Western blot and chromatin immunoprecipitation (ChIP) were used to explore the role of Ikzf2 on ICOS expression. Results After S. japonicum infection, the expression of ICOS molecules gradually increased in splenic lymphocytes, especially in Th cells (P < 0.01). Compared with ICOS- Th cells, more ICOS+ Th cells expressed CD69, CD25, CXCR5, and CD40L (P < 0.05), while less of them expressed CD62L (P < 0.05). Also, ICOS+ Th cells expressed more cytokines, such as IFN-γ, IL-4, IL-10, IL-2, and IL-21 (P < 0.05). RNA sequencing results showed that many transcription factors were increased significantly in ICOS+ Th cells, especially Ikzf2 (P < 0.05). And then, the expression of Ikzf2 was verified to be significantly increased and mainly located in the nuclear of ICOS+ Th cells. Finally, ChIP experiments and dual-luciferase reporter assay confirmed that Ikzf2 could directly bind to the ICOS promoter in Th cells. Conclusion In this study, ICOS+ Th cells were found to play an important role in S. japonicum infection to induce immune response in the spleen of C57BL/6 mice. Additionally, Ikzf2 was found to be one important transcription factor that could regulate the expression of ICOS in the spleen of S. japonicum-infected C57BL/6 mice.
Collapse
Affiliation(s)
- Shihao Xie
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haixia Wei
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Anping Peng
- Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Anqi Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiajie Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao Fang
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Feihu Shi
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Quan Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - He Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongyan Xie
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xingfei Pan
- Department of Infectious Diseases, Key Laboratory for Major Obsteric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xu Tian
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- College of Pharmacy, Guangzhou Medical University, Guangzhou, China
| | - Jun Huang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
49
|
Nunes-Santos CJ, Kuehn HS, Rosenzweig SD. IKAROS Family Zinc Finger 1-Associated Diseases in Primary Immunodeficiency Patients. Immunol Allergy Clin North Am 2021; 40:461-470. [PMID: 32654692 DOI: 10.1016/j.iac.2020.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ikaros zinc finger 1 (IKZF1 or Ikaros) is a hematopoietic zinc finger DNA-binding transcription factor that acts as a critical regulator of lymphocyte and myeloid differentiation. Loss-of-function germline heterozygous mutations in IKZF1 affecting DNA-binding were described as causative of 2 distinct primary immunodeficiency (PID)/inborn error of immunity diseases. Mutations acting by haploinsufficiency present with a common variable immune deficiency-like phenotype mainly characterized by increased susceptibility to infections. Mutations acting in a dominant negative fashion present with a combined immunodeficiency phenotype with high prevalence of Pneumocystis jirovecii pneumonia. Pathophysiology and manifestations of IKAROS-associated diseases in patients with PID are reviewed here.
Collapse
Affiliation(s)
- Cristiane J Nunes-Santos
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, 10 Center Drive, Building 10, Room 2C410F, Bethesda, MD 20892, USA
| | - Hye Sun Kuehn
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, 10 Center Drive, Building 10, Room 2C410F, Bethesda, MD 20892, USA
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, National Institutes of Health (NIH) Clinical Center, 10 Center Drive, Building 10, Room 2C410F, Bethesda, MD 20892, USA.
| |
Collapse
|
50
|
Wang ES, Verano AL, Nowak RP, Yuan JC, Donovan KA, Eleuteri NA, Yue H, Ngo KH, Lizotte PH, Gokhale PC, Gray NS, Fischer ES. Acute pharmacological degradation of Helios destabilizes regulatory T cells. Nat Chem Biol 2021; 17:711-717. [PMID: 34035522 PMCID: PMC8162940 DOI: 10.1038/s41589-021-00802-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/19/2021] [Indexed: 02/02/2023]
Abstract
The zinc-finger transcription factor Helios is critical for maintaining the identity, anergic phenotype and suppressive activity of regulatory T (Treg) cells. While it is an attractive target to enhance the efficacy of currently approved immunotherapies, no existing approaches can directly modulate Helios activity or abundance. Here, we report the structure-guided development of small molecules that recruit the E3 ubiquitin ligase substrate receptor cereblon to Helios, thereby promoting its degradation. Pharmacological Helios degradation destabilized the anergic phenotype and reduced the suppressive activity of Treg cells, establishing a route towards Helios-targeting therapeutics. More generally, this study provides a framework for the development of small-molecule degraders for previously unligandable targets by reprogramming E3 ligase substrate specificity.
Collapse
Affiliation(s)
- Eric S. Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Alyssa L. Verano
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Radosław P. Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - J. Christine Yuan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Katherine A. Donovan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | | | - Hong Yue
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Kenneth H. Ngo
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA
| | - Patrick H. Lizotte
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA
| | - Prafulla C. Gokhale
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA.,Correspondence to: Nathanael S. Gray (); Eric S. Fischer ()
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA.,Correspondence to: Nathanael S. Gray (); Eric S. Fischer ()
| |
Collapse
|