151
|
Calreticulin mutations affect its chaperone function and perturb the glycoproteome. Cell Rep 2022; 41:111689. [DOI: 10.1016/j.celrep.2022.111689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/17/2022] [Accepted: 10/27/2022] [Indexed: 11/23/2022] Open
|
152
|
Many faces of SF3B1-mutated myeloid neoplasms: concurrent mutational profiles contribute to the diverse clinical and morphologic features. Hum Pathol 2022; 129:81-89. [PMID: 36087739 DOI: 10.1016/j.humpath.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 12/14/2022]
Abstract
Splicing factor SF3B1 mutation occurs in 20-30% of myelodysplastic syndrome (MDS) and myelodysplasia/myeloproliferative neoplasm (MDS/MPN), particularly those with ring sideroblasts (RS), and rarely in acute myeloid leukemia (AML). In this study, we performed a comprehensive evaluation of 77 SF3B1-mutated myeloid neoplasms (45 MDS, 18 MDS/MPN, 13 AML, and 1 MPN), including their clinical presentations, morphologic features, cytogenetic studies, and targeted next-generation sequencing. Our study demonstrated that concurrent gene mutations were very different in SF3B1-mutated MDS, MDS/MPN, and AML. MDS cases were frequently characterized by either sole SF3B1 mutation or in combination with TET2 mutation. Acquiring additional mutations in transcription factors, such as RUNX1 and GATA2, were associated with increased blasts and progression to AML in patients with MDS or MDS/MPN. Our study also demonstrated that SF3B1-mutated MDS/MPN was not only associated with thrombocytosis (5/18, 27.7%), defined by the current WHO classification as MDS/MPN-RS-T, but also associated with neutrophilia (6/18, 33.3%), monocytosis (6/18, 33.3%), and mastocytosis (1/18, 5.6%). Our results indicate that although SF3B1-mutated myeloid neoplasms in general have a good prognosis, evaluation of the concurrent gene mutational profile is important for risk stratification. In addition, our study, in combination with other published data, suggests that the category of MDS/MPN-RS-T in the current WHO classification could be expanded to include SF3B1-mutated MDS/MPN-RS with peripheral leukocytosis such as neutrophilia and monocytosis.
Collapse
|
153
|
Kim HY, Han Y, Jang JH, Jung CW, Kim SH, Kim HJ. Effects of CALR-Mutant Type and Burden on the Phenotype of Myeloproliferative Neoplasms. Diagnostics (Basel) 2022; 12:2570. [PMID: 36359414 PMCID: PMC9689478 DOI: 10.3390/diagnostics12112570] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 08/13/2023] Open
Abstract
Somatic CALR mutations occur in approximately 70% of patients with JAK2 V617F-negative essential thrombocythemia (ET) and primary myelofibrosis (PMF). We evaluated the effects of the CALR mutant type and burden on the phenotype of CALR-mutated myeloproliferative neoplasms (MPN). Of the 510 patients with suspected or diagnosed MPN, all 49 patients detected with CALR mutations were diagnosed with ET (n = 32) or PMF (n = 17). The CALR mutant burden was significantly higher in PMF than in ET (45% vs. 34%), and type 1-like and type 2-like mutations were detected in 49% and 51% patients, respectively. Patients with MPN and type 2-like mutation showed a significantly higher median platelet count than those with type 1-like mutation. Particularly, patients with ET and type 2-like mutation had no thrombotic events, despite higher platelet counts. The effect of CALR mutant burden differed depending on the mutant type. A higher mutant burden tended to be associated with a cytopenic phenotype (i.e., lower hemoglobin levels and platelet counts) in patients with the type 1-like mutation and a proliferative hematological phenotype (i.e., higher platelet and neutrophil counts) in patients with the type 2-like mutation. This study suggests that the disease phenotype of MPN may be altered through CALR mutant burden and mutant type.
Collapse
Affiliation(s)
- Hyun-Young Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Yujin Han
- Department of Laboratory Medicine, Seegene Medical Foundation, Seoul 04805, Korea
| | - Jun Ho Jang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Chul Won Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Sun-Hee Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| |
Collapse
|
154
|
Clonogenic assays improve determination of variant allele frequency of driver mutations in myeloproliferative neoplasms. Ann Hematol 2022; 101:2655-2663. [PMID: 36269400 DOI: 10.1007/s00277-022-05000-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/01/2022] [Indexed: 11/27/2022]
Abstract
Molecular diagnostics moves more into focus as technology advances. In patients with myeloproliferative neoplasms (MPN), identification and monitoring of the driver mutations have become an integral part of diagnosis and monitoring of the disease. In some patients, none of the known driver mutations (JAK2V617F, CALR, MPL) is found, and they are termed "triple negative" (TN). Also, whole-blood variant allele frequency (VAF) of driver mutations may not adequately reflect the VAF in the stem cells driving the disease. We reasoned that colony forming unit (CFU) assay-derived clonogenic cells may be better suited than next-generation sequencing (NGS) of whole blood to detect driver mutations in TN patients and to provide a VAF of disease-driving cells. We have included 59 patients carrying the most common driver mutations in the establishment or our model. Interestingly, cloning efficiency correlated with whole blood VAF (p = 0.0048), suggesting that the number of disease-driving cells correlated with VAF. Furthermore, the clonogenic VAF correlated significantly with the NGS VAF (p < 0.0001). This correlation was lost in patients with an NGS VAF <15%. Further analysis showed that in patients with a VAF <15% by NGS, clonogenic VAF was higher than NGS VAF (p = 0.003), suggesting an enrichment of low numbers of disease-driving cells in CFU assays. However, our approach did not enhance the identification of driver mutations in 5 TN patients. A significant correlation of lactate dehydrogenase (LDH) serum levels with both CFU- and NGS-derived VAF was found. Our results demonstrate that enrichment for clonogenic cells can improve the detection of MPN driver mutations in patients with low VAF and that LDH levels correlate with VAF.
Collapse
|
155
|
Sun M, Qi S, Wu M, Xia W, Xiong H. Calreticulin as a prognostic biomarker and correlated with immune infiltrate in kidney renal clear cell carcinoma. Front Genet 2022; 13:909556. [PMID: 36338983 PMCID: PMC9633671 DOI: 10.3389/fgene.2022.909556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 10/05/2022] [Indexed: 01/29/2024] Open
Abstract
Background: Calreticulin (CALR) has been investigated in several malignant diseases and is associated with immune-cell infiltration. However, the prognostic value of CALR in kidney renal clear cell carcinoma (KIRC) is still unknown. Methods: Based on the computational analysis, data from 530 KIRC cases and 72 normal kidney samples from The Cancer Genome Atlas (TGCA-KIRC) database were analyzed in this study. The expression of CALR mRNA in pan-cancer and immune infiltrates was analyzed using the Tumor Immune Estimation Resource (TIMER) database. The CALR protein expression was obtained from the UALCAN and Human Protein Atlas (HPA) databases. Survival, functional, and statistical analyses were conducted using R software. Results: The CALR expression was higher in KIRC cases than in normal kidneys. A high CALR expression was correlated with TNM stage, pathological stage, and histological grade. Kaplan-Meier survival analysis showed that a high CALR expression was associated with poor overall survival, disease-specific survival, and progression-free interval. Gene set enrichment analysis (GSEA) indicated that CALR was enriched in IL-6 and IL-2 signaling, interferon signaling, TNF signaling, inflammatory response, apoptosis, and the p53 pathway. CALR is correlated with immune-infiltrating cells. A significant correlation was observed between CALR expression and immunomodulators. Conclusion: We identified CALR as a prognostic biomarker of KIRC. Meanwhile, the CALR expression associated with immune infiltration indicated that CALR might be a potential immunotherapy target for patients with KIRC.
Collapse
Affiliation(s)
| | | | | | | | - Hao Xiong
- Department of Hematology and Oncology, Wuhan Children’s Hospital, Tongji Medical College, HUST, Wuhan, China
| |
Collapse
|
156
|
Paes J, Silva GAV, Tarragô AM, Mourão LPDS. The Contribution of JAK2 46/1 Haplotype in the Predisposition to Myeloproliferative Neoplasms. Int J Mol Sci 2022; 23:12582. [PMID: 36293440 PMCID: PMC9604447 DOI: 10.3390/ijms232012582] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 11/17/2022] Open
Abstract
Haplotype 46/1 (GGCC) consists of a set of genetic variations distributed along chromosome 9p.24.1, which extend from the Janus Kinase 2 gene to Insulin like 4. Marked by four jointly inherited variants (rs3780367, rs10974944, rs12343867, and rs1159782), this haplotype has a strong association with the development of BCR-ABL1-negative myeloproliferative neoplasms (MPNs) because it precedes the acquisition of the JAK2V617F variant, a common genetic alteration in individuals with these hematological malignancies. It is also described as one of the factors that increases the risk of familial MPNs by more than five times, 46/1 is associated with events related to inflammatory dysregulation, splenomegaly, splanchnic vein thrombosis, Budd-Chiari syndrome, increases in RBC count, platelets, leukocytes, hematocrit, and hemoglobin, which are characteristic of MPNs, as well as other findings that are still being elucidated and which are of great interest for the etiopathological understanding of these hematological neoplasms. Considering these factors, the present review aims to describe the main findings and discussions involving the 46/1 haplotype, and highlights the molecular and immunological aspects and their relevance as a tool for clinical practice and investigation of familial cases.
Collapse
Affiliation(s)
- Jhemerson Paes
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil
| | - George A. V. Silva
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (FHEMOAM), Manaus 69050-001, AM, Brazil
- Fundação Oswaldo Cruz–Instituto Leônidas e Maria Deane (Fiocruz), Manaus 69027-070, AM, Brazil
| | - Andréa M. Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (FHEMOAM), Manaus 69050-001, AM, Brazil
| | - Lucivana P. de Souza Mourão
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (FHEMOAM), Manaus 69050-001, AM, Brazil
| |
Collapse
|
157
|
Immanuel T, Li J, Green TN, Bogdanova A, Kalev-Zylinska ML. Deregulated calcium signaling in blood cancer: Underlying mechanisms and therapeutic potential. Front Oncol 2022; 12:1010506. [PMID: 36330491 PMCID: PMC9623116 DOI: 10.3389/fonc.2022.1010506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Intracellular calcium signaling regulates diverse physiological and pathological processes. In solid tumors, changes to calcium channels and effectors via mutations or changes in expression affect all cancer hallmarks. Such changes often disrupt transport of calcium ions (Ca2+) in the endoplasmic reticulum (ER) or mitochondria, impacting apoptosis. Evidence rapidly accumulates that this is similar in blood cancer. Principles of intracellular Ca2+ signaling are outlined in the introduction. We describe different Ca2+-toolkit components and summarize the unique relationship between extracellular Ca2+ in the endosteal niche and hematopoietic stem cells. The foundational data on Ca2+ homeostasis in red blood cells is discussed, with the demonstration of changes in red blood cell disorders. This leads to the role of Ca2+ in neoplastic erythropoiesis. Then we expand onto the neoplastic impact of deregulated plasma membrane Ca2+ channels, ER Ca2+ channels, Ca2+ pumps and exchangers, as well as Ca2+ sensor and effector proteins across all types of hematologic neoplasms. This includes an overview of genetic variants in the Ca2+-toolkit encoding genes in lymphoid and myeloid cancers as recorded in publically available cancer databases. The data we compiled demonstrate that multiple Ca2+ homeostatic mechanisms and Ca2+ responsive pathways are altered in hematologic cancers. Some of these alterations may have genetic basis but this requires further investigation. Most changes in the Ca2+-toolkit do not appear to define/associate with specific disease entities but may influence disease grade, prognosis, treatment response, and certain complications. Further elucidation of the underlying mechanisms may lead to novel treatments, with the aim to tailor drugs to different patterns of deregulation. To our knowledge this is the first review of its type in the published literature. We hope that the evidence we compiled increases awareness of the calcium signaling deregulation in hematologic neoplasms and triggers more clinical studies to help advance this field.
Collapse
Affiliation(s)
- Tracey Immanuel
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jixia Li
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Department of Laboratory Medicine, School of Medicine, Foshan University, Foshan City, China
| | - Taryn N. Green
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| | - Maggie L. Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
- Haematology Laboratory, Department of Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
158
|
Mughal TI, Pemmaraju N, Bejar R, Gale RP, Bose P, Kiladjian JJ, Prchal J, Royston D, Pollyea D, Valent P, Brümmendorf TH, Skorski T, Patnaik M, Santini V, Fenaux P, Kucine N, Verstovsek S, Mesa R, Barbui T, Saglio G, Van Etten RA. Perspective: Pivotal translational hematology and therapeutic insights in chronic myeloid hematopoietic stem cell malignancies. Hematol Oncol 2022; 40:491-504. [PMID: 35368098 DOI: 10.1002/hon.2987] [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: 12/28/2021] [Revised: 02/21/2022] [Accepted: 03/03/2022] [Indexed: 11/10/2022]
Abstract
Despite much of the past 2 years being engulfed by the devastating consequences of the SAR-CoV-2 pandemic, significant progress, even breathtaking, occurred in the field of chronic myeloid malignancies. Some of this was show-cased at the 15th Post-American Society of Hematology (ASH) and the 25th John Goldman workshops on myeloproliferative neoplasms (MPN) held on 9th-10th December 2020 and 7th-10th October 2021, respectively. The inaugural Post-ASH MPN workshop was set out in 2006 by John Goldman (deceased) and Tariq Mughal to answer emerging translational hematology and therapeutics of patients with these malignancies. Rather than present a resume of the discussions, this perspective focuses on some of the pivotal translational hematology and therapeutic insights in these diseases.
Collapse
Affiliation(s)
- Tariq I Mughal
- Tufts University School of Medicine, Boston, Massachusetts, USA
- University of Buckingham, Buckingham, UK
| | - Naveen Pemmaraju
- MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Rafael Bejar
- University of California San Diego, La Jolla, California, USA
| | | | - Prithviraj Bose
- MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | | | - Josef Prchal
- Huntsman Cancer Center, Salt Lake City, Utah, USA
| | - Daniel Royston
- John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Daniel Pollyea
- University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Peter Valent
- Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Tomasz Skorski
- Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Valeria Santini
- Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Pierre Fenaux
- Hospital St Louis, Assistance Publique Hôpitaux de Paris, Paris, France
| | | | - Srdan Verstovsek
- MD Anderson Cancer Center, University of Texas, Houston, Texas, USA
| | - Ruben Mesa
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, Texas, USA
| | - Tiziano Barbui
- Fondazione per la Ricerca Ospedale Maggiore di Bergamo, Bergamo, Italy
| | | | - Richard A Van Etten
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California, USA
| |
Collapse
|
159
|
Polyclonal evolution of Fanconi anemia to MDS and AML revealed at single cell resolution. Exp Hematol Oncol 2022; 11:64. [PMID: 36167633 PMCID: PMC9513989 DOI: 10.1186/s40164-022-00319-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
Background Fanconi anemia (FA) is a rare disease of bone marrow failure. FA patients are prone to develop myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). However, the molecular clonal evolution of the progression from FA to MDS/AML remains elusive. Methods Herein, we performed a comprehensive genomic analysis using an FA patient (P1001) sample that transformed to MDS and subsequently AML, together with other three FA patient samples at the MDS stage. Results Our finding showed the existence of polyclonal pattern in these cases at MDS stage. The clonal evolution analysis of FA case (P1001) showed the mutations of UBASH3A, SF3B1, RUNX1 and ASXL1 gradually appeared at the later stage of MDS, while the IDH2 alteration become the dominant clone at the leukemia stage. Moreover, single-cell sequencing analyses further demonstrated a polyclonal pattern was present at either MDS or AML stages, whereas IDH2 mutated cell clones appeared only at the leukemia stage. Conclusions We thus propose a clonal evolution model from FA to MDS and AML for this patient. The results of our study on the clonal evolution and mutated genes of the progression of FA to AML are conducive to understanding the progression of the disease that still perplexes us. Supplementary Information The online version contains supplementary material available at 10.1186/s40164-022-00319-5.
Collapse
|
160
|
Weller MC, Haralambieva E, Bühler MM, Benz R, Theocharides APA, Balabanov S. CALR loss-of-heterozygosity as a potential driver for extramedullary AML. Ann Hematol 2022; 101:2571-2573. [PMID: 36155839 DOI: 10.1007/s00277-022-04976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/30/2022] [Indexed: 11/01/2022]
Affiliation(s)
- Marie-Christine Weller
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Eugenia Haralambieva
- Department of Pathology and Molecular Pathology, University Hospital Zurich and University of Zurich, Zurich, Switzerland.,Institute for Clinical Pathology Medica, Hottingerstrasse 9/11, 8032, Zurich, Switzerland
| | - Marco Matteo Bühler
- Department of Pathology and Molecular Pathology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Rudolf Benz
- Division of Hematology and Oncology, Kantonsspital Muensterlingen, Muensterlingen, Switzerland
| | - Alexandre Pierre André Theocharides
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Stefan Balabanov
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
161
|
Clinical Features, Gene Alterations, and Outcomes in Prefibrotic and Overt Primary and Secondary Myelofibrotic Patients. Cancers (Basel) 2022; 14:cancers14184485. [PMID: 36139644 PMCID: PMC9496754 DOI: 10.3390/cancers14184485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/02/2022] [Accepted: 09/10/2022] [Indexed: 11/17/2022] Open
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs) are divided in three major groups: polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The 2016 WHO classification incorporates also prefibrotic PMF (pre-PMF) and overt PMF. This study aimed to discriminate the clinical features, genetic alterations, and outcomes in patients with prefibrotic, overt PMF, and secondary MF (SMF). This study included 229 patients with diagnosed myelofibrosis (MF). Among 229 patients, 67 (29%), 122 (53%), and 40 (18%) were confirmed as SMF, overt PMF, and pre-PMF, respectively. The JAK2 V617F mutation was differentially distributed in SMF and PMF, contradictory to CALR and MPL mutations. Regarding nondriver mutations, the occurrence of ASXL1 mutations differed between PMF and SMF or pre-PMF. The three-year overall survival was 91.5%, 85.3%, and 94.8% in SMF, overt PMF, and pre-PMF groups. Various scoring systems could discriminate the overall survival in PMF but not in SMF and pre-PMF. Still, clinical features including anemia and thrombocytopenia were poor prognostic factors throughout the myelofibrosis, whereas mutations contributed differently. Molecular grouping by wild-type SF3B1 and SRSF2/RUNX1/U2AF1/ASXL1/TP53 mutations showed inferior progression-free survival (PFS) in PMF, SMF, and pre-PMF. We determined the clinical and genetic features related to poor prognosis in myelofibrosis.
Collapse
|
162
|
Mutant CALR's "sweet tooth". Blood 2022; 140:1187-1189. [PMID: 36107460 PMCID: PMC9479035 DOI: 10.1182/blood.2022017448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 11/20/2022] Open
|
163
|
Jutzi JS, Marneth AE, Ciboddo M, Guerra-Moreno A, Jiménez-Santos MJ, Kosmidou A, Dressman JW, Liang H, Hamel R, Lozano P, Rumi E, Doench JG, Gotlib J, Krishnan A, Elf S, Al-Shahrour F, Mullally A. Whole-genome CRISPR screening identifies N-glycosylation as a genetic and therapeutic vulnerability in CALR-mutant MPNs. Blood 2022; 140:1291-1304. [PMID: 35763665 PMCID: PMC9479036 DOI: 10.1182/blood.2022015629] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 06/10/2022] [Indexed: 01/13/2023] Open
Abstract
Calreticulin (CALR) mutations are frequent, disease-initiating events in myeloproliferative neoplasms (MPNs). Although the biological mechanism by which CALR mutations cause MPNs has been elucidated, there currently are no clonally selective therapies for CALR-mutant MPNs. To identify unique genetic dependencies in CALR-mutant MPNs, we performed a whole-genome clustered regularly interspaced short palindromic repeats (CRISPR) knockout depletion screen in mutant CALR-transformed hematopoietic cells. We found that genes in the N-glycosylation pathway (among others) were differentially depleted in mutant CALR-transformed cells as compared with control cells. Using a focused pharmacological in vitro screen targeting unique vulnerabilities uncovered in the CRISPR screen, we found that chemical inhibition of N-glycosylation impaired the growth of mutant CALR-transformed cells, through a reduction in MPL cell surface expression. We treated Calr-mutant knockin mice with the N-glycosylation inhibitor 2-deoxy-glucose (2-DG) and found a preferential sensitivity of Calr-mutant cells to 2-DG as compared with wild-type cells and normalization of key MPNs disease features. To validate our findings in primary human cells, we performed megakaryocyte colony-forming unit (CFU-MK) assays. We found that N-glycosylation inhibition significantly reduced CFU-MK formation in patient-derived CALR-mutant bone marrow as compared with bone marrow derived from healthy donors. In aggregate, our findings advance the development of clonally selective treatments for CALR-mutant MPNs.
Collapse
Affiliation(s)
- Jonas S Jutzi
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anna E Marneth
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michele Ciboddo
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL
| | - Angel Guerra-Moreno
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - María José Jiménez-Santos
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Anastasia Kosmidou
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - James W Dressman
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC
| | - Hongyan Liang
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC
| | - Rebecca Hamel
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- RWTH Aachen University, Aachen, Germany
| | - Patricia Lozano
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elisa Rumi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Hematology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, Pavia, Italy
| | | | - Jason Gotlib
- Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Anandi Krishnan
- Department of Pathology, Stanford Cancer Institute, Stanford University School of Medicine, Palo Alto, CA; and
| | - Shannon Elf
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- The Ben May Department for Cancer Research, University of Chicago, Chicago, IL
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Structural Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Broad Institute, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
164
|
Inferring the initiation and development of myeloproliferative neoplasms. Proc Natl Acad Sci U S A 2022; 119:e2120374119. [PMID: 36083966 PMCID: PMC9478641 DOI: 10.1073/pnas.2120374119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The developmental history of blood cancer begins with mutation acquisition and the resulting malignant clone expansion. The two most prevalent driver mutations found in myeloproliferative neoplasms-JAK2V617F and CALRm-occur in hematopoietic stem cells, which are highly complex to observe in vivo. To circumvent this difficulty, we propose a method relying on mathematical modeling and statistical inference to determine disease initiation and dynamics. Our findings suggest that CALRm mutations tend to occur later in life than JAK2V617F. Our results confirm the higher proliferative advantage of the CALRm malignant clone compared to JAK2V617F. Furthermore, we illustrate how mathematical modeling and Bayesian inference can be used for setting up early screening strategies.
Collapse
|
165
|
Brown DW, Zhou W, Wang Y, Jones K, Luo W, Dagnall C, Teshome K, Klein A, Zhang T, Lin SH, Lee OW, Khan S, Vo JB, Hutchinson A, Liu J, Wang J, Zhu B, Hicks B, Martin AS, Spellman SR, Wang T, Deeg HJ, Gupta V, Lee SJ, Freedman ND, Yeager M, Chanock SJ, Savage SA, Saber W, Gadalla SM, Machiela MJ. Germline-somatic JAK2 interactions are associated with clonal expansion in myelofibrosis. Nat Commun 2022; 13:5284. [PMID: 36075929 PMCID: PMC9458655 DOI: 10.1038/s41467-022-32986-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022] Open
Abstract
Myelofibrosis is a rare myeloproliferative neoplasm (MPN) with high risk for progression to acute myeloid leukemia. Our integrated genomic analysis of up to 933 myelofibrosis cases identifies 6 germline susceptibility loci, 4 of which overlap with previously identified MPN loci. Virtual karyotyping identifies high frequencies of mosaic chromosomal alterations (mCAs), with enrichment at myelofibrosis GWAS susceptibility loci and recurrently somatically mutated MPN genes (e.g., JAK2). We replicate prior MPN associations showing germline variation at the 9p24.1 risk haplotype confers elevated risk of acquiring JAK2V617F mutations, demonstrating with long-read sequencing that this relationship occurs in cis. We also describe recurrent 9p24.1 large mCAs that selectively retained JAK2V617F mutations. Germline variation associated with longer telomeres is associated with increased myelofibrosis risk. Myelofibrosis cases with high-frequency JAK2 mCAs have marked reductions in measured telomere length - suggesting a relationship between telomere biology and myelofibrosis clonal expansion. Our results advance understanding of the germline-somatic interaction at JAK2 and implicate mCAs involving JAK2 as strong promoters of clonal expansion of those mutated clones.
Collapse
Affiliation(s)
- Derek W Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA.
| | - Weiyin Zhou
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Youjin Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Wen Luo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Casey Dagnall
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Kedest Teshome
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Shu-Hong Lin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Olivia W Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Sairah Khan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Jacqueline B Vo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Jia Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Jiahui Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Andrew St Martin
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program, Minneapolis, MN, USA
| | - Tao Wang
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - H Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Vikas Gupta
- MPN Program, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Stephanie J Lee
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory, Frederick, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Wael Saber
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shahinaz M Gadalla
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| |
Collapse
|
166
|
Gerds AT, Gotlib J, Ali H, Bose P, Dunbar A, Elshoury A, George TI, Gundabolu K, Hexner E, Hobbs GS, Jain T, Jamieson C, Kaesberg PR, Kuykendall AT, Madanat Y, McMahon B, Mohan SR, Nadiminti KV, Oh S, Pardanani A, Podoltsev N, Rein L, Salit R, Stein BL, Talpaz M, Vachhani P, Wadleigh M, Wall S, Ward DC, Bergman MA, Hochstetler C. Myeloproliferative Neoplasms, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022; 20:1033-1062. [PMID: 36075392 DOI: 10.6004/jnccn.2022.0046] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) consist of myelofibrosis, polycythemia vera, and essential thrombocythemia and are a heterogeneous group of clonal blood disorders characterized by an overproduction of blood cells. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for MPN were developed as a result of meetings convened by a multidisciplinary panel with expertise in MPN, with the goal of providing recommendations for the management of MPN in adults. The Guidelines include recommendations for the diagnostic workup, risk stratification, treatment, and supportive care strategies for the management of myelofibrosis, polycythemia vera, and essential thrombocythemia. Assessment of symptoms at baseline and monitoring of symptom status during the course of treatment is recommended for all patients. This article focuses on the recommendations as outlined in the NCCN Guidelines for the diagnosis of MPN and the risk stratification, management, and supportive care relevant to MF.
Collapse
Affiliation(s)
- Aaron T Gerds
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | - Haris Ali
- City of Hope National Medical Center
| | | | | | | | | | | | | | | | - Tania Jain
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | | | | | | - Stephen Oh
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | - Rachel Salit
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | - Sarah Wall
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Dawn C Ward
- UCLA Jonsson Comprehensive Cancer Center; and
| | | | | |
Collapse
|
167
|
Arai K, Sakaguchi M, Yui S, Kitano T, Miyata M, Yogosawa M, Nakayama K, Tajika K, Usuki K, Kuroda J, Uoshima N, Kobayashi Y, Uchiyama H, Kubota Y, Kimura S, Mori S, Hirai M, Wakita S, Yamaguchi H. Simultaneous detection of JAK2, CALR, and MPL mutations and quantitation of JAK2 V617F allele burden in myeloproliferative neoplasms using the quenching probe-Tm method in i-densy IS-5320. Int J Lab Hematol 2022; 44:1102-1110. [PMID: 36039795 DOI: 10.1111/ijlh.13938] [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: 02/12/2022] [Accepted: 07/07/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Accurate detection of myeloproliferative neoplasms (MPN)-associated gene mutations is necessary to correctly diagnose MPN. However, conventional gene testing has various limitations, including the requirement of skilled technicians, cumbersome experimental procedures, and turnaround time of several days. The gene analyzer i-densy IS-5320 allows gene testing using the quenching probe-Tm method. Specifically, pretreatment of samples including DNA extraction, amplification and detection of genes, and analysis of results are performed in a fully automatic manner after samples and test reagents are added into this system, which is compact and can be easily installed in a laboratory. The aim of this study is to investigate the sensitivity and specificity associated with the simultaneous detection of MPN-associated gene mutations. METHODS We conducted an analysis of MPN-associated genes using i-densy IS-5320. We analyzed 384 samples (171 JAK2 V617F mutations, 10 JAK2 exon12 mutations, 104 CALR mutations, and 26 MPL mutations) that had been examined using conventional approaches such as allele-specific polymerase chain reaction (PCR), droplet digital PCR, and the direct sequencing method. RESULTS The detection accuracy of JAK2 V617F, JAK2 exon 12, CALR, and MPL was 100.0% (383/383), 99.7% (383/384), 100.0% (370/370), and 99.7% (377/378), respectively. There was a strong positive correlation between the JAK2 V617F allele burden measured using conventional methods and i-densy IS-5320 (r = .989). CONCLUSION Overall, i-densy IS-5320 exhibited good accuracy in terms of analyzing MPN-associated genes; thus, it can serve as a replacement for conventional methods of MPN-associated gene testing.
Collapse
Affiliation(s)
- Kunihito Arai
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | | | - Shunsuke Yui
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Tomoaki Kitano
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Miho Miyata
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Mayumi Yogosawa
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Kazutaka Nakayama
- Department of Hematology, Yokohama Minami Kyousai Hospital, Kanagawa, Japan
| | - Kenji Tajika
- Department of Hematology, Yokohama Minami Kyousai Hospital, Kanagawa, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Nobuhiko Uoshima
- Department of Hematology, Japanese Red Cross, Kyoto Daini Hospital, Kyoto, Japan
| | - Yutaka Kobayashi
- Department of Hematology, Japanese Red Cross, Kyoto Daini Hospital, Kyoto, Japan
| | - Hitoji Uchiyama
- Department of Hematology, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Yasushi Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Shinichiro Mori
- Hemato-Oncology Department, St Luke's International Hospital, Tokyo, Japan
| | | | - Satoshi Wakita
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
168
|
Kapeni C, Nitsche L, Kilpatrick AM, Wilson NK, Xia K, Mirshekar-Syahkal B, Chandrakanthan V, Malouf C, Pimanda JE, Göttgens B, Kirschner K, Tomlinson SR, Ottersbach K. p57Kip2 regulates embryonic blood stem cells by controlling sympathoadrenal progenitor expansion. Blood 2022; 140:464-477. [PMID: 35653588 PMCID: PMC9353151 DOI: 10.1182/blood.2021014853] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/13/2022] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are of major clinical importance, and finding methods for their in vitro generation is a prime research focus. We show here that the cell cycle inhibitor p57Kip2/Cdkn1c limits the number of emerging HSCs by restricting the size of the sympathetic nervous system (SNS) and the amount of HSC-supportive catecholamines secreted by these cells. This regulation occurs at the SNS progenitor level and is in contrast to the cell-intrinsic function of p57Kip2 in maintaining adult HSCs, highlighting profound differences in cell cycle requirements of adult HSCs compared with their embryonic counterparts. Furthermore, this effect is specific to the aorta-gonad-mesonephros (AGM) region and shows that the AGM is the main contributor to early fetal liver colonization, as early fetal liver HSC numbers are equally affected. Using a range of antagonists in vivo, we show a requirement for intact β2-adrenergic signaling for SNS-dependent HSC expansion. To gain further molecular insights, we have generated a single-cell RNA-sequencing data set of all Ngfr+ sympathoadrenal cells around the dorsal aorta to dissect their differentiation pathway. Importantly, this not only defined the relevant p57Kip2-expressing SNS progenitor stage but also revealed that some neural crest cells, upon arrival at the aorta, are able to take an alternative differentiation pathway, giving rise to a subset of ventrally restricted mesenchymal cells that express important HSC-supportive factors. Neural crest cells thus appear to contribute to the AGM HSC niche via 2 different mechanisms: SNS-mediated catecholamine secretion and HSC-supportive mesenchymal cell production.
Collapse
Affiliation(s)
- Chrysa Kapeni
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Leslie Nitsche
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola K Wilson
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kankan Xia
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Bahar Mirshekar-Syahkal
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Vashe Chandrakanthan
- School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Camille Malouf
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - John E Pimanda
- School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- Department of Haematology, The Prince of Wales Hospital, Sydney, NSW, Australia
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kristina Kirschner
- Institute of Cancer Sciences and
- CRUK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom
| | - Simon R Tomlinson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Katrin Ottersbach
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
169
|
Todisco G, Moura PL, Hellström-Lindberg E. Clinical manifestations of clonal hematopoiesis: What has SF3B1-mutant MDS taught us? Semin Hematol 2022; 59:150-155. [DOI: 10.1053/j.seminhematol.2022.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 12/17/2022]
|
170
|
Aoyama R, Kubota Y, Tara S, Wakita S, Yamaguchi H, Shimizu W, Takano H. Vascular Endothelial Dysfunction in Myeloproliferative Neoplasms and Gene Mutations. Int Heart J 2022; 63:661-668. [PMID: 35831151 DOI: 10.1536/ihj.22-003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Essential thrombocythemia (ET) and polycythemia vera (PV), are common Philadelphia-negative myeloproliferative neoplasms (MPN). Patients with MPN have a high rate of cardiovascular complications and often have acquired JAK2V617F and CALR genetic mutations. In this study, we aimed to analyze vascular endothelial function in patients with MPN.We evaluated 27 outpatients, including 10 patients diagnosed with MPN, flow-mediated dilatation (FMD), and nitroglycerin-mediated dilation (NMD), between September 2014 and August 2016. We measured serum adiponectin, which protects vascular endothelial function, and serum asymmetric dimethyl arginine (ADMA), which inhibits the production of adiponectin. The presence or absence of JAK2V617F and CALR mutations was evaluated in patients with MPN.Venous thrombosis was observed more frequently in patients with MPN than in those without. Seven MPN patients were diagnosed with PV, and 3 MPN patients were diagnosed with ET. JAK2V617F and CALR mutations were found in 5 and 3 MPN patients, respectively. FMD was significantly lower in JAK2V617F-positive MPN patients than in JAK2V617F-negative MPN patients, although NMD, adiponectin, and ADMA were similar in both groups. Adiponectin levels were higher and ADMA levels were lower in CALR-positive MPN patients than in CALR-negative MPN patients. There was no difference in FMD and NMD prevalence between the 2 groups. Furthermore, we had 3 representative MPN patients who were complicated with coronary spasm, possibly caused by MPN-related endothelial dysfunction.We found that patients with MPN presented with endothelial dysfunction, which was related to the presence of genetic mutations and was sometimes associated with cardiovascular disease.
Collapse
Affiliation(s)
- Rie Aoyama
- Department of Cardiovascular Medicine, Nippon Medical School.,Division of Cardiology, Heart and Vascular Institute, Funabashi Municipal Medical Center
| | - Yoshiaki Kubota
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Shuhei Tara
- Department of Cardiovascular Medicine, Nippon Medical School
| | | | | | - Wataru Shimizu
- Department of Cardiovascular Medicine, Nippon Medical School
| | - Hitoshi Takano
- Department of Cardiovascular Medicine, Nippon Medical School
| |
Collapse
|
171
|
Campbell AG, Seelig DM, Beckman JD, Minor KM, Heinrich DA, Friedenberg SG, Modiano JF, Furrow E. Targeted sequencing of candidate gene regions for myelofibrosis in dogs. J Vet Intern Med 2022; 36:1237-1247. [PMID: 35815881 PMCID: PMC9308436 DOI: 10.1111/jvim.16476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 06/12/2022] [Indexed: 12/31/2022] Open
Abstract
Background Myelofibrosis often lacks an identifiable cause in dogs. In humans, most primary myelofibrosis cases develop secondary to driver mutations in JAK2, CALR, or MPL. Objectives To determine the prevalence of variants in JAK2, CALR, or MPL candidate regions in dogs with myelofibrosis and in healthy dogs. Animals Twenty‐six dogs with myelofibrosis that underwent bone marrow biopsy between 2010 and 2018 and 25 control dogs matched for age, sex, and breed. Methods Cross‐sectional study. Amplicon sequencing of JAK2 exons 12 and 14, CALR exon 9, and MPL exon 10 was performed on formalin‐fixed, decalcified, paraffin‐embedded bone marrow (myelofibrosis) or peripheral blood (control) DNA. Somatic variants were categorized as likely‐benign or possibly‐pathogenic based on predicted impact on protein function. Within the myelofibrosis group, hematologic variables and survival were compared by variant status (none, likely‐benign only, and ≥1 possibly‐pathogenic). The effect of age on variant count was analyzed using linear regression. Results Eighteen of 26 (69%) myelofibrosis cases had somatic variants, including 9 classified as possibly‐pathogenic. No somatic variants were detected in controls. Within the myelofibrosis group, hematologic variables and survival did not differ by variant status. The number of somatic variants per myelofibrosis case increased with age (estimate, 0.69; SE, 0.29; P = .03). Conclusions and Clinical Importance Somatic variants might initiate or perpetuate myelofibrosis in dogs. Our findings suggest the occurrence of clonal hematopoiesis in dogs, with increasing incidence with age, as observed in humans.
Collapse
Affiliation(s)
- Amelia G Campbell
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Davis M Seelig
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Joan D Beckman
- Division of Hematology, Oncology and Transplantation, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Katie M Minor
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Daniel A Heinrich
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota, USA
| | - Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota, USA.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA.,Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eva Furrow
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.,Animal Cancer Care and Research Program, University of Minnesota, St. Paul, Minnesota, USA
| |
Collapse
|
172
|
Edahiro Y, Ito T, Gotoh A, Nakamae M, Kimura F, Koike M, Kirito K, Wada H, Usuki K, Tanaka T, Mori T, Wakita S, Saito TI, Kada A, Saito AM, Shimoda K, Sugimoto Y, Kurokawa T, Tomita A, Hashimoto Y, Akashi K, Matsumura I, Takenaka K, Komatsu N. Clinical characteristics of Japanese patients with polycythemia vera: results of the JSH-MPN-R18 study. Int J Hematol 2022; 116:696-711. [PMID: 35809214 DOI: 10.1007/s12185-022-03412-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
Abstract
The presence of a JAK2 V617F or JAK2 exon 12 mutation is one of the three major criteria listed for the diagnosis of polycythemia vera (PV) in the 2017 World Health Organization Classification. However, a nationwide study has not yet been conducted in Japan since the discovery of JAK2 mutations. Therefore, the Japanese Society of Hematology (JSH) retrospectively analyzed the clinical characteristics of 596 Japanese patients with PV diagnosed between April 2005 and March 2018. Among the 473 patients with complete data on JAK2 mutations available, 446 (94.3%) and 10 (2.1%) were positive for the JAK2 V617F and JAK2 exon 12 mutations, respectively. During a median follow-up of 46 months (range: 0-179 months), 47 (7.9%) deaths occurred. The major causes of death were secondary malignancies (23.4%), acute leukemia (12.8%), non-leukemic progressive disease (10.6%) and thrombotic (6.4%) and hemorrhagic complications (6.4%). Thrombotic and hemorrhagic events occurred during the clinical course in 4.0% (n = 24) and 3.5% (n = 21) of patients, respectively. These results show that the international PV prognostic score (age, venous thrombosis and leukocytosis) is applicable to Japanese patients with PV.
Collapse
Affiliation(s)
- Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.,Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Akihiko Gotoh
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Mika Nakamae
- Hematology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Fumihiko Kimura
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Michiaki Koike
- Department of Hematology, Juntendo University Shizuoka Hospital, Shizuoka, Japan
| | - Keita Kirito
- Department of Hematology and Oncology, University of Yamanashi, Yamanashi, Japan
| | - Hideho Wada
- Department of Hematology, Kawasaki Medical School, Okayama, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Takayuki Tanaka
- Department of Hematology, Tottori Prefectural Central Hospital, Tottori, Japan
| | - Takehiko Mori
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Department of Hematology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Wakita
- Division of Hematology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Toshiki I Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Akiko Kada
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yuka Sugimoto
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Mie, Japan
| | - Toshiro Kurokawa
- Department of Hematology, Toyama Red Cross Hospital, Toyama, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Yoshinori Hashimoto
- Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Hematology, Tottori Prefectural Central Hospital, Tottori, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Itaru Matsumura
- Division of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan. .,Laboratory for the Development of Therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,PharmaEssentia Japan KK, Tokyo, Japan.
| |
Collapse
|
173
|
Real world study of children and young adults with myeloproliferative neoplasms identifying risks and unmet needs. Blood Adv 2022; 6:5171-5183. [PMID: 35802458 PMCID: PMC9631631 DOI: 10.1182/bloodadvances.2022007201] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
In a contemporary cohort of 444 young MPN patients, risks of thrombosis, hemorrhage, and transformation were 1% pt/y. Current risk scores had no utility. Uniquely, we identify that splenomegaly and hyperviscosity symptoms predict thrombosis and transformation.
Myeloproliferative neoplasms (MPNs) are uncommon in children/young adults. Here, we present data on unselected patients diagnosed before 25 years of age included from 38 centers in 15 countries. Sequential patients were included. We identified 444 patients, with median follow-up 9.7 years (0-47.8). Forty-nine (11.1%) had a history of thrombosis at diagnosis, 49 new thrombotic events were recorded (1.16% patient per year [pt/y]), perihepatic vein thromboses were most frequent (47.6% venous events), and logistic regression identified JAK2V617F mutation (P = .016) and hyperviscosity symptoms (visual disturbances, dizziness, vertigo, headache) as risk factors (P = .040). New hemorrhagic events occurred in 44 patients (9.9%, 1.04% pt/y). Disease transformation occurred in 48 patients (10.9%, 1.13% pt/y), usually to myelofibrosis (7.5%) with splenomegaly as a novel risk factor for transformation in essential thrombocythemia (ET) (P= .000) in logistical regression. Eight deaths (1.8%) were recorded, 3 after allogeneic stem cell transplantation. Concerning conventional risk scores: International Prognostic Score for Essential Thrombocythemia-Thrombosis and new International Prognostic Score for Essential Thrombocythemia-Thrombosis differentiated ET patients in terms of thrombotic risk. Both scores identified high-risk patients with the same median thrombosis-free survival of 28.5 years. No contemporary scores were able to predict survival for young ET or polycythemia vera patients. Our data represents the largest real-world study of MPN patients age < 25 years at diagnosis. Rates of thrombotic events and transformation were higher than expected compared with the previous literature. Our study provides new and reliable information as a basis for prospective studies, trials, and development of harmonized international guidelines for the specific management of young patients with MPN.
Collapse
|
174
|
Murakami S, Barroca V, Perié L, Bravard A, Bernardino-Sgherri J, Tisserand A, Devanand C, Edmond V, Magniez A, Tenreira Bento S, Torres C, Pasquier F, Plo I, Vainchenker W, Villeval JL, Roméo PH, Lewandowski D. In Vivo Monitoring of Polycythemia Vera Development Reveals Carbonic Anhydrase 1 as a Potent Therapeutic Target. Blood Cancer Discov 2022; 3:285-297. [PMID: 35290450 PMCID: PMC9327731 DOI: 10.1158/2643-3230.bcd-21-0039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/21/2021] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Current murine models of myeloproliferative neoplasms (MPNs) cannot examine how MPNs progress from a single bone marrow source to the entire hematopoietic system. Thus, using transplantation of knock-in JAK2V617F hematopoietic cells into a single irradiated leg, we show development of polycythemia vera (PV) from a single anatomic site in immunocompetent mice. Barcode experiments reveal that grafted JAK2V617F stem/progenitor cells migrate from the irradiated leg to nonirradiated organs such as the contralateral leg and spleen, which is strictly required for development of PV. Mutant cells colonizing the nonirradiated leg efficiently induce PV in nonconditioned recipient mice and contain JAK2V617F hematopoietic stem/progenitor cells that express high levels of carbonic anhydrase 1 (CA1), a peculiar feature also found in CD34+ cells from patients with PV. Finally, genetic and pharmacologic inhibition of CA1 efficiently suppresses PV development and progression in mice and decreases PV patients' erythroid progenitors, strengthening CA1 as a potent therapeutic target for PV. SIGNIFICANCE Follow-up of hematopoietic malignancies from their initiating anatomic site is crucial for understanding their development and discovering new therapeutic avenues. We developed such an approach, used it to characterize PV progression, and identified CA1 as a promising therapeutic target of PV. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Shohei Murakami
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | - Vilma Barroca
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | - Leïla Perié
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | - Anne Bravard
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | - Jacqueline Bernardino-Sgherri
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | | | - Caroline Devanand
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | - Valérie Edmond
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Aurélie Magniez
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | | | - Claire Torres
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | | | - Isabelle Plo
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | | | | | - Paul-Henri Roméo
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| | - Daniel Lewandowski
- CEA-INSERM UMR1274, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), CEA, Fontenay aux Roses, France
- Université Paris Diderot, Paris, France
- Université Paris Saclay, Gif-sur-Yvette, France
| |
Collapse
|
175
|
Ibarra J, Elbanna YA, Kurylowicz K, Ciboddo M, Greenbaum HS, Arellano NS, Rodriguez D, Evers M, Bock-Hughes A, Liu C, Smith Q, Lutze J, Baumeister J, Kalmer M, Olschok K, Nicholson B, Silva D, Maxwell L, Dowgielewicz J, Rumi E, Pietra D, Casetti IC, Catricala S, Koschmieder S, Gurbuxani S, Schneider RK, Oakes SA, Elf SE. Type I but Not Type II Calreticulin Mutations Activate the IRE1α/XBP1 Pathway of the Unfolded Protein Response to Drive Myeloproliferative Neoplasms. Blood Cancer Discov 2022; 3:298-315. [PMID: 35405004 PMCID: PMC9338758 DOI: 10.1158/2643-3230.bcd-21-0144] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/21/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023] Open
Abstract
Approximately 20% of patients with myeloproliferative neoplasms (MPN) harbor mutations in the gene calreticulin (CALR), with 80% of those mutations classified as either type I or type II. While type II CALR-mutant proteins retain many of the Ca2+ binding sites present in the wild-type protein, type I CALR-mutant proteins lose these residues. The functional consequences of this differential loss of Ca2+ binding sites remain unexplored. Here, we show that the loss of Ca2+ binding residues in the type I mutant CALR protein directly impairs its Ca2+ binding ability, which in turn leads to depleted endoplasmic reticulum (ER) Ca2+ and subsequent activation of the IRE1α/XBP1 pathway of the unfolded protein response. Genetic or pharmacologic inhibition of IRE1α/XBP1 signaling induces cell death in type I mutant but not type II mutant or wild-type CALR-expressing cells, and abrogates type I mutant CALR-driven MPN disease progression in vivo. SIGNIFICANCE Current targeted therapies for CALR-mutated MPNs are not curative and fail to differentiate between type I- versus type II-driven disease. To improve treatment strategies, it is critical to identify CALR mutation type-specific vulnerabilities. Here we show that IRE1α/XBP1 represents a unique, targetable dependency specific to type I CALR-mutated MPNs. This article is highlighted in the In This Issue feature, p. 265.
Collapse
Affiliation(s)
- Juan Ibarra
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Yassmin A. Elbanna
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Katarzyna Kurylowicz
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Michele Ciboddo
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Harrison S. Greenbaum
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Nicole S. Arellano
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Deborah Rodriguez
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Maria Evers
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Althea Bock-Hughes
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Chenyu Liu
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Quinn Smith
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Julian Lutze
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, Illinois
| | - Julian Baumeister
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Milena Kalmer
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Kathrin Olschok
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Benjamin Nicholson
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| | - Diane Silva
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Luke Maxwell
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
| | - Jonathan Dowgielewicz
- Committee on Molecular Metabolism and Nutrition, University of Chicago, Chicago, Illinois
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | | | - Silvia Catricala
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University and Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | | | - Rebekka K. Schneider
- Department of Cell Biology, Institute for Biomedical Technologies, RWTH Aachen University, Aachen, Germany
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Scott A. Oakes
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Shannon E. Elf
- The Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois
- Committee on Cancer Biology, University of Chicago, Chicago, Illinois
| |
Collapse
|
176
|
Alzahrani M, Al Turki S, Al Rajban W, Alshalati F, Almodaihsh F, Abuelgasim KA, Alahmari B, Al Bogami T, Ali O, Al Harbi T, AlBalwi MA, Alotaibi M, Aleem A, Al Asker A, Al Mugairi A. Pro106Leu MPL mutation is associated with thrombocytosis and a low risk of thrombosis, splenomegaly and marrow fibrosis. Platelets 2022; 33:1220-1227. [PMID: 35791502 DOI: 10.1080/09537104.2022.2091773] [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: 10/17/2022]
Abstract
The P106L mutation in the human myeloproliferative leukemia virus oncogene (MPL) was shown to be associated with hereditary thrombocythemia in Arabs. The clinical and bone marrow (BM) features of P106L mutation are unknown. Genetic databases at two tertiary hospitals in Saudi Arabia were searched to identify patients with the MPL P106L mutation. Clinical data were collected retrospectively and the BM aspirates and biopsies were independently reviewed by two hematopathologists. In total, 115 patients were included. Median age was 33 years of which 31 patients were pediatric and 65 were female. The mutation was homozygous in 87 patients. Thrombocytosis was documented in 107 patients, with a median platelet count of 667 × 109/L. The homozygous genotype was associated with a higher platelet count. Thirty-three patients had an evaluable BM and clustering of megakaryocytes was observed in 30/33 patients. At the time of last follow-up, 114 patients were alive. The median follow-up was 7.8 years from the time of thrombocytosis. No patients developed disease progression to myelofibrosis. The P106L mutation was associated with marked thrombocytosis at a younger age and with a low risk of thrombosis, splenomegaly, and marrow fibrosis. The BM demonstrated normal or hypocellular marrow with megakaryocyte clusters.
Collapse
Affiliation(s)
- Musa Alzahrani
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Saeed Al Turki
- Department of Pathology and Laboratory Medicine, Molecular Pathology Division, King Abdulaziz Medical City, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Waleed Al Rajban
- Department of Pathology and Laboratory Medicine, Molecular Pathology Division, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Fatimah Alshalati
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Fahad Almodaihsh
- Department of Pathology and Laboratory Medicine, Hematopathology Unit, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Khadega A Abuelgasim
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,Division of Adult Hematology and Stem Cell Transplant, Department of Oncology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Bader Alahmari
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,Division of Adult Hematology and Stem Cell Transplant, Department of Oncology, King Abdulaziz Medical City, Riyadh, Saudi Arabia.,Department of Oncology, Ministry of the National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Thamer Al Bogami
- Department of Pathology and Laboratory Medicine, Hematopathology Unit, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Osama Ali
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,Division of Adult Hematology and Stem Cell Transplant, Department of Oncology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Talal Al Harbi
- Department of Pediatric Hematology and Oncology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Mohammed A AlBalwi
- Department of Pathology and Laboratory Medicine, Molecular Pathology Division, King Abdulaziz Medical City, Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Maram Alotaibi
- Department of Pathology and Laboratory Medicine, Molecular Genetics Unit, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Aamer Aleem
- Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed Al Asker
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,Division of Adult Hematology and Stem Cell Transplant, Department of Oncology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Areej Al Mugairi
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,Department of Pathology and Laboratory Medicine, Hematopathology Division, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
177
|
Saha C, Harrison C. Fedratinib, the first selective JAK2 inhibitor approved for treatment of myelofibrosis - an option beyond ruxolitinib. Expert Rev Hematol 2022; 15:583-595. [PMID: 35787092 DOI: 10.1080/17474086.2022.2098105] [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: 11/04/2022]
Abstract
Introduction: Myelofibrosis, a life shortening clonal disorder, presents with a constellation of features: bone marrow fibrosis, abnormal blood counts, extramedullary hematopoiesis, splenomegaly, thrombohemorrhagic complications and constitutional symptoms. Until recently Ruxolitinib, a JAK1 and 2 inhibitor, has been the only targeted therapy available for transplant-ineligible patients requiring treatment for splenomegaly and disease related symptoms. However, most patients discontinue Ruxolitinib after 3-5 years, mostly due to loss of response. There has been an unmet need for this patient group. In August 2019 Fedratinib (INREBIC® capsules, Impact Biomedicines, Inc., a wholly owned subsidiary of Bristol Meyer Squibb), a JAK2 inhibitor, was approved by US FDA for treatment of myelofibrosis in both JAK inhibitor naïve and pre-treated patients for the management of symptoms and splenomegaly.Areas covered: Here, we discuss the development, evidence base to date for Fedratinib. Including early and late phase, and ongoing trials, safety issues, potential role and current position of Fedratinib in the treatment of myelofibrosis, as well as future direction of targeted therapy in myelofibrosis.Expert opinion: Fedratinib presents a much needed option of treatment, particularly, for patients failing Ruxolitinib, with response rates that are quite similar. Nonetheless, there remain important questions including sequencing and options for combining therapy.
Collapse
Affiliation(s)
- Chandan Saha
- Department of Hematology, Guy's and St Thomas' NHS Foundation Trust, London
| | - Claire Harrison
- Department of Hematology, Guy's and St Thomas' NHS Foundation Trust, London
| |
Collapse
|
178
|
Gerds AT, Lyons RM, Colucci P, Kalafut P, Paranagama D, Verstovsek S. Disease and Clinical Characteristics of Patients With a Clinical Diagnosis of Myelofibrosis Enrolled in the MOST Study. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e532-e540. [PMID: 35256316 DOI: 10.1016/j.clml.2022.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Clinical characteristics and treatment patterns of patients with lower-risk myelofibrosis (MF) are not well described. This analysis from the MOST (NCT02953704) assessed the demographic and clinical characteristics and treatment patterns of patients with the clinical diagnosis of lower-risk MF at enrollment. PATIENTS AND METHODS MOST is an ongoing, prospective, observational study in patients with clinical diagnoses of MF or essential thrombocythemia enrolled at clinical practices throughout the United States. Patients included in the MF cohort (≥18 years of age) had low-risk MF by the Dynamic International Prognostic Scoring System or intermediate-1 (INT-1) risk MF (by age >65 years only) at enrollment. Patient data were entered into an electronic case report form during usual-care visits over a planned 36 month observation period. RESULTS Two hundred five patients were eligible for this analysis (low risk, n = 85; INT-1 risk, n = 120; median age, 68 years [range, 35-88]); 166 patients (81.0%) had mutation testing results available. The median time from MF diagnosis to enrollment was 1.8 years. Hemoglobin and hematocrit levels were below the normal range in 50.5% and 48.7% of patients, respectively. Nearly all (98.0%) patients had comorbid conditions, most commonly hypertension (49.8%). Fatigue was the most common physician-reported MF symptom (30.7%). At enrollment, 55.6% of patients were receiving MF-directed monotherapy, most frequently hydroxyurea (46.5%) or ruxolitinib (40.4%). CONCLUSION Future longitudinal analyses of data from MOST will help identify unmet needs and characterize how patients with lower-risk MF are managed throughout the disease course.
Collapse
Affiliation(s)
- Aaron T Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH.
| | - Roger M Lyons
- Texas Oncology and US Oncology Research, San Antonio, TX
| | | | | | | | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
179
|
Abdelghani M, Hammami H, Zidi W, Amouri H, Othmen HBH, Farrah A, Menif S. Hematological relevance of JAK2 V617F and calreticulin mutations in Tunisian patients with essential thrombocythemia. J Clin Lab Anal 2022; 36:e24522. [PMID: 35754115 PMCID: PMC9396186 DOI: 10.1002/jcla.24522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 04/28/2022] [Accepted: 05/01/2022] [Indexed: 11/14/2022] Open
Abstract
Background The genetic investigation of essential thrombocythemia(ET) has highlighted the presence of driver mutations in ET. Janus kinase JAK2V617F and calreticulin(CALR) mutations are the most frequent driver mutations and have significantly improved the molecular diagnosis of ET. The impact of genetic heterogeneity on clinical features has not been fully elucidated. This is the first study which aimed to determine the frequency of JAK2V617F and CALR exon9 mutations in Tunisian ET patients and to establish the correlation between hematological characteristics and mutational status. Methods This study included Tunisian patients suspected with ET and was conducted between September 2017 and March 2021. Genomic DNA of patients was isolated from peripheral blood samples. JAK2V617F was detected by AS‐PCR and CALR mutations were detected by PCR/direct sequencing. Clinical and hematological characteristics were also analyzed. Results Two hundred and fifty ET patients were enrolled in this study. JAK2V617F mutation was found in 166/250 (66.4%) of patients, whereas CALR mutations were detected in 27/84 (32.1%) patients without JAK2V617F. Compared with JAK2V617F‐positive patients, those with CALR mutations showed lower hemoglobin level and lower leucocytes count (p = 0.007 and p = 0.004,respectively). CALR type 2 was the most frequent mutation of CALR detected in 55.55% of CALR mutated. Six of seven patients with thrombotic events harbored JAK2V617F mutation. Conclusion The prevalence of driver mutations JAK2V617F or CALR mutations was 77.2% in Tunisian ET patients. Moreover, patients with JAK2 V617F mutation had a higher risk of thrombosis. The mutational status is necessary to improve the diagnosis and contribute to the therapeutic decisions.
Collapse
Affiliation(s)
- Maroua Abdelghani
- LR16IPT07, Molecular and Cellular Hematology Laboratory, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Faculty of Mathematics, Physics and Natural Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Haifa Hammami
- LR16IPT07, Molecular and Cellular Hematology Laboratory, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia.,Faculty of Mathematics, Physics and Natural Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Wiem Zidi
- Laboratory of Biochemistry, Rabta Hospital, Tunis, Tunisia
| | - Hassiba Amouri
- LR16IPT07, Molecular and Cellular Hematology Laboratory, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Hind Ben Hadj Othmen
- LR16IPT07, Molecular and Cellular Hematology Laboratory, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Ahlem Farrah
- LR16IPT07, Molecular and Cellular Hematology Laboratory, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Samia Menif
- LR16IPT07, Molecular and Cellular Hematology Laboratory, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
180
|
Theranostic Potentials of Gold Nanomaterials in Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14133047. [PMID: 35804818 PMCID: PMC9264814 DOI: 10.3390/cancers14133047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/03/2022] [Accepted: 06/17/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hematological malignancies (HMs) cover 50% of all malignancies, and people of all ages can be affected by these deadly diseases. In many cases, conventional diagnostic tools fail to diagnose HMs at an early stage, due to heterogeneity and the long-term indolent phase of HMs. Therefore, many patients start their treatment at the late stage of HMs and have poor survival. Gold nanomaterials (GNMs) have shown promise as a cancer theranostic agent. GNMs are 1 nm to 100 nm materials having magnetic resonance and surface-plasmon-resonance properties. GNMs conjugated with antibodies, nucleic acids, peptides, photosensitizers, chemotherapeutic drugs, synthetic-drug candidates, bioactive compounds, and other theranostic biomolecules may enhance the efficacy and efficiency of both traditional and advanced theranostic approaches to combat HMs. Abstract Hematological malignancies (HMs) are a heterogeneous group of blood neoplasia generally characterized by abnormal blood-cell production. Detection of HMs-specific molecular biomarkers (e.g., surface antigens, nucleic acid, and proteomic biomarkers) is crucial in determining clinical states and monitoring disease progression. Early diagnosis of HMs, followed by an effective treatment, can remarkably extend overall survival of patients. However, traditional and advanced HMs’ diagnostic strategies still lack selectivity and sensitivity. More importantly, commercially available chemotherapeutic drugs are losing their efficacy due to adverse effects, and many patients develop resistance against these drugs. To overcome these limitations, the development of novel potent and reliable theranostic agents is urgently needed to diagnose and combat HMs at an early stage. Recently, gold nanomaterials (GNMs) have shown promise in the diagnosis and treatment of HMs. Magnetic resonance and the surface-plasmon-resonance properties of GNMs have made them a suitable candidate in the diagnosis of HMs via magnetic-resonance imaging and colorimetric or electrochemical sensing of cancer-specific biomarkers. Furthermore, GNMs-based photodynamic therapy, photothermal therapy, radiation therapy, and targeted drug delivery enhanced the selectivity and efficacy of anticancer drugs or drug candidates. Therefore, surface-tuned GNMs could be used as sensitive, reliable, and accurate early HMs, metastatic HMs, and MRD-detection tools, as well as selective, potent anticancer agents. However, GNMs may induce endothelial leakage to exacerbate cancer metastasis. Studies using clinical patient samples, patient-derived HMs models, or healthy-animal models could give a precise idea about their theranostic potential as well as biocompatibility. The present review will investigate the theranostic potential of vectorized GNMs in HMs and future challenges before clinical theranostic applications in HMs.
Collapse
|
181
|
Production and Characterization of Peptide Antibodies to the C-Terminal of Frameshifted Calreticulin Associated with Myeloproliferative Diseases. Int J Mol Sci 2022; 23:ijms23126803. [PMID: 35743246 PMCID: PMC9223637 DOI: 10.3390/ijms23126803] [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: 04/07/2022] [Revised: 06/04/2022] [Accepted: 06/15/2022] [Indexed: 11/25/2022] Open
Abstract
Myeloproliferative Neoplasms (MPNs) constitute a group of rare blood cancers that are characterized by mutations in bone marrow stem cells leading to the overproduction of erythrocytes, leukocytes, and thrombocytes. Mutations in calreticulin (CRT) genes may initiate MPNs, causing a novel variable polybasic stretch terminating in a common C-terminal sequence in the frameshifted CRT (CRTfs) proteins. Peptide antibodies to the mutated C-terminal are important reagents for research in the molecular mechanisms of MPNs and for the development of new diagnostic assays and therapies. In this study, eight peptide antibodies targeting the C-terminal of CRTfs were produced and characterised by modified enzyme-linked immunosorbent assays using resin-bound peptides. The antibodies reacted to two epitopes: CREACLQGWTE for SSI-HYB 385-01, 385-02, 385-03, 385-04, 385-07, 385-08, and 385-09 and CLQGWT for SSI-HYB 385-06. For the majority of antibodies, the residues Cys1, Trp9, and Glu11 were essential for reactivity. SSI-HYB 385-06, with the highest affinity, recognised recombinant CRTfs produced in yeast and the MARIMO cell line expressing CRTfs when examined in Western immunoblotting. Moreover, SSI-HYB 385-06 occasionally reacted to CRTfs from MPN patients when analysed by flow cytometry. The characterized antibodies may be used to understand the role of CRTfs in the pathogenesis of MPNs and to design and develop new diagnostic assays and therapeutic targets.
Collapse
|
182
|
Ali EA, Abu-Tineh M, Rozi W, Ali B, Babiker A, Hailan Y, Al-Maharmeh Q, Maat Z, Ismail A, Yassin MA. The Outcome of Fatherhood in Patients With Philadelphia-Negative Myeloproliferative Neoplasms: A Single-Institution Experience. Cureus 2022; 14:e25953. [PMID: 35855233 PMCID: PMC9286008 DOI: 10.7759/cureus.25953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2022] [Indexed: 11/22/2022] Open
Abstract
Background Fertility is a highly complex subject; it involves more than one individual and has profound psychological and economic implications. Moreover, it is affected by several factors, including age, significant systemic illness in either partner, exposure to environmental toxins, medications, or radiation. In patients with malignancy, fertility is more complicated. Patients with a malignancy might have reduced fertility due to the disease, medication, and radiation. Besides the reduced fertility, there are more concerns regarding the subsequent effect of cancer treatment on their offspring and the possibility of having healthy children. There were many studies regarding fertility in patients with cancer; however, in male patients with Philadelphia-negative myeloproliferative neoplasms (MPNs), there are very limited data. Objectives In this study, we aim to see the outcome of fatherhood in male patients with Philadelphia-negative myeloproliferative neoplasms (MPNs), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) whether on treatment or not. Methods A retrospective mixed-design study of male patients with Philadelphia-negative MPN was followed up in our institute (National Center for Cancer Care and Research (NCCCR)), Doha, Qatar, between January 1, 2008, and January 1, 2020. Patients were interviewed regarding fertility-related information. All included patients had a confirmed diagnosis of Philadelphia-negative MPN according to World Health Organization (WHO) 2008 or WHO 2016 criteria for MPN, aged more than 18 years old. Results A total of 124 male patients were interviewed, and only 20 patients met the inclusion criteria. The majority of the patients were lost to follow-up or could not be contacted, and 28.8% of the excluded patients had their families completed by the time of diagnosis. The treatment received included hydroxycarbamide (n=8), pegylated interferon 2 alpha (n=10), ruxolitinib (n=1), and phlebotomy (n=1). The mean duration of exposure to treatment before pregnancy was 4.7 years. The mode of delivery was normal vaginal delivery in 71.4% of the pregnancies. The total number of offspring was 30, and the total number of conceptions was 30. Conclusion Our data showed that most Philadelphia-negative MPN male patients on treatment had their offspring born normally with no serious complications, congenital anomalies, or reports of MPN-related cancers. Patients’ concerns regarding fertility should be addressed well to ensure a better quality of life.
Collapse
|
183
|
Gigoux M, Holmström MO, Zappasodi R, Park JJ, Pourpe S, Bozkus CC, Mangarin LMB, Redmond D, Verma S, Schad S, George MM, Venkatesh D, Ghosh A, Hoyos D, Molvi Z, Kamaz B, Marneth AE, Duke W, Leventhal MJ, Jan M, Ho VT, Hobbs GS, Knudsen TA, Skov V, Kjær L, Larsen TS, Hansen DL, Lindsley RC, Hasselbalch H, Grauslund JH, Lisle TL, Met Ö, Wilkinson P, Greenbaum B, Sepulveda MA, Chan T, Rampal R, Andersen MH, Abdel-Wahab O, Bhardwaj N, Wolchok JD, Mullally A, Merghoub T. Calreticulin mutant myeloproliferative neoplasms induce MHC-I skewing, which can be overcome by an optimized peptide cancer vaccine. Sci Transl Med 2022; 14:eaba4380. [PMID: 35704596 PMCID: PMC11182673 DOI: 10.1126/scitranslmed.aba4380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The majority of JAK2V617F-negative myeloproliferative neoplasms (MPNs) have disease-initiating frameshift mutations in calreticulin (CALR), resulting in a common carboxyl-terminal mutant fragment (CALRMUT), representing an attractive source of neoantigens for cancer vaccines. However, studies have shown that CALRMUT-specific T cells are rare in patients with CALRMUT MPN for unknown reasons. We examined class I major histocompatibility complex (MHC-I) allele frequencies in patients with CALRMUT MPN from two independent cohorts. We observed that MHC-I alleles that present CALRMUT neoepitopes with high affinity are underrepresented in patients with CALRMUT MPN. We speculated that this was due to an increased chance of immune-mediated tumor rejection by individuals expressing one of these MHC-I alleles such that the disease never clinically manifested. As a consequence of this MHC-I allele restriction, we reasoned that patients with CALRMUT MPN would not efficiently respond to a CALRMUT fragment cancer vaccine but would when immunized with a modified CALRMUT heteroclitic peptide vaccine approach. We found that heteroclitic CALRMUT peptides specifically designed for the MHC-I alleles of patients with CALRMUT MPN efficiently elicited a CALRMUT cross-reactive CD8+ T cell response in human peripheral blood samples but not to the matched weakly immunogenic CALRMUT native peptides. We corroborated this effect in vivo in mice and observed that C57BL/6J mice can mount a CD8+ T cell response to the CALRMUT fragment upon immunization with a CALRMUT heteroclitic, but not native, peptide. Together, our data emphasize the therapeutic potential of heteroclitic peptide-based cancer vaccines in patients with CALRMUT MPN.
Collapse
Affiliation(s)
- Mathieu Gigoux
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Morten O. Holmström
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY 10021, USA
| | - Joseph J. Park
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Stephane Pourpe
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Levi M. B. Mangarin
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Redmond
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Svena Verma
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Sara Schad
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Mariam M. George
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Divya Venkatesh
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Arnab Ghosh
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zaki Molvi
- Weill Cornell Medicine, New York, NY 10065, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Baransel Kamaz
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anna E. Marneth
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - William Duke
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Max Jan
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Vincent T. Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela S. Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Trine Alma Knudsen
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | | | - Dennis Lund Hansen
- Department of Hematology, Odense University Hospital, Odense 5000, Denmark
| | - R. Coleman Lindsley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Hans Hasselbalch
- Department of Hematology, Zealand University Hospital, Roskilde 4000, Denmark
| | - Jacob H. Grauslund
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Thomas L. Lisle
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Özcan Met
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Patrick Wilkinson
- Janssen Oncology Therapeutic Area, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19002, USA
| | - Benjamin Greenbaum
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medicine, Physiology, Biophysics and Systems Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Manuel A. Sepulveda
- Janssen Oncology Therapeutic Area, Janssen Research and Development, LLC, Pharmaceutical Companies of Johnson & Johnson, Spring House, PA 19002, USA
| | - Timothy Chan
- Weill Cornell Medical College, New York, NY 10065, USA
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raajit Rampal
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mads H. Andersen
- Department of Oncology, National Center for Cancer Immune Therapy, Herlev Hospital, Herlev 2730, Denmark
- Department of Immunology and Microbiology, Copenhagen University Hospital, Herlev 2730, Denmark
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program and Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nina Bhardwaj
- Parker Institute for Cancer Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jedd D. Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Ann Mullally
- Department of Medicine, Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute, Cambridge, MA 02142, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Human Oncology and Pathogenesis Program and Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
184
|
Florez MA, Tran BT, Wathan TK, DeGregori J, Pietras EM, King KY. Clonal hematopoiesis: Mutation-specific adaptation to environmental change. Cell Stem Cell 2022; 29:882-904. [PMID: 35659875 PMCID: PMC9202417 DOI: 10.1016/j.stem.2022.05.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) describes a widespread expansion of genetically variant hematopoietic cells that increases exponentially with age and is associated with increased risks of cancers, cardiovascular disease, and other maladies. Here, we discuss how environmental contexts associated with CHIP, such as old age, infections, chemotherapy, or cigarette smoking, alter tissue microenvironments to facilitate the selection and expansion of specific CHIP mutant clones. Further, we consider major remaining gaps in knowledge, including intrinsic effects, clone size thresholds, and factors affecting clonal competition, that will determine future application of this field in transplant and preventive medicine.
Collapse
Affiliation(s)
- Marcus A Florez
- Medical Scientist Training Program and Program in Translational Biology and Molecular Medicine, Graduate School of Biomedical Sciences, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA; Division of Infectious Disease, Department of Pediatrics, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA
| | - Brandon T Tran
- Graduate School of Biomedical Sciences, Program in Cancer and Cell Biology, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA; Division of Infectious Disease, Department of Pediatrics, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA
| | - Trisha K Wathan
- Division of Infectious Disease, Department of Pediatrics, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Microbiology and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric M Pietras
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Microbiology and Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine Y King
- Medical Scientist Training Program and Program in Translational Biology and Molecular Medicine, Graduate School of Biomedical Sciences, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA; Graduate School of Biomedical Sciences, Program in Cancer and Cell Biology, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA; Division of Infectious Disease, Department of Pediatrics, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, 1102 Bates Street, Suite 1150, Houston, TX 77030, USA.
| |
Collapse
|
185
|
Clinical impact of glycans in platelet and megakaryocyte biology. Blood 2022; 139:3255-3263. [PMID: 35015813 PMCID: PMC9164739 DOI: 10.1182/blood.2020009303] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/23/2021] [Indexed: 11/20/2022] Open
Abstract
Humans produce and remove 1011 platelets daily to maintain a steady-state platelet count. The tight regulation of platelet production and removal from the blood circulation prevents anomalies in both processes from resulting in reduced or increased platelet count, often associated with the risk of bleeding or overt thrombus formation, respectively. This review focuses on the role of glycans, also known as carbohydrates or oligosaccharides, including N- and O-glycans, proteoglycans, and glycosaminoglycans, in human and mouse platelet and megakaryocyte physiology. Based on recent clinical observations and mouse models, we focused on the pathologic aspects of glycan biosynthesis and degradation and their effects on platelet numbers and megakaryocyte function.
Collapse
|
186
|
Wang B, Wen L, Wang Z, Chen S, Qiu H. Differential Implications of CSF3R Mutations in t(8;21) and CEBPA Double Mutated Acute Myeloid Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:393-404. [PMID: 34975010 DOI: 10.1016/j.clml.2021.11.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Few data are available exploring mutations of the colony-stimulating factor 3 receptor (CSF3R) in acute myeloid leukemia (AML) in an all-round and systematic manner. The purpose of this study was to analyze the CSF3R mutations (CSF3Rmut) in AML with recurrent genetic abnormalities for potential synergistic pathomechanism. PATIENTS AND METHODS We retrospectively screened 1102 adult de novo AML patients with available next-generation sequencing (NGS) information on 132 genes related to hematologic disorders. The χ2, Mann-Whitney U tests were used to analyze their associations with clinicopathologic characteristics, and a propensity score matching (PSM) followed by Kaplan-Meier method was applied to measure their prognostic effects. RESULTS Overall, CSF3Rmut were detected in 40 (3.6%) of 1102 patients with adult de novo AML. CSF3Rmut were predominantly enriched in AML with the CEBPA double mutations (CEBPAdm) (16/122, 13.1%), t(8;21) (12/186, 6.5%) and mutated RUNX1 (3/50, 6.0%), respectively. The CSF3Rmut loci and types differed according to AML subtypes, with frameshift-indels and premature stop confined in the t(8;21) AML [10/12 (83.3%)], and missense recurrently aggregated in the CEBPAdm AML [16/16 (100%)]. Cases with CSF3Rmut had a lower WBC count versus those with CSF3R wild-type (CSF3Rwt) in the t(8;21) AML cohort, with a borderline significance [median 5.45 (range 0.94-20.30) × 109/L) vs. 8.80 (range 0.96-155.00) × 109/L, P = .046]. CSF3Rmut were non-significantly associated with higher WBC counts [median 33.6 (range 6.8-287.6) × 109/L vs. 18.1 (range 1.7-196.0) × 109/L, P = .156] and significantly with lower immunophenotypic CD15 positivity [0/8 (0%) vs. 44/80 (55%), P = .009] as compared to CSF3Rwt in the CEBPAdm AML cohort. After propensity score matching followed by Kaplan-Meier analysis, CSF3Rmut cases had comparable disease-free survival (DFS) and overall survival (OS) to those with CSF3Rwt (P = .607 and P = .842, respectively) in the t(8;21) AML cohort. By contrast, CSF3Rmut showed an inclination towards inferior DFS compared to CSF3Rwt in the CEBPAdm AML cohort [median DFS 19.8 (95%CI 3.1-36.5) months vs. not reached (NR), P = .086]. No significant difference was found for OS between CSF3Rmut and CSF3Rwt cases (P = .943). CONCLUSION We concluded that CSF3Rmut were frequently enriched in patients with t(8;21) and CEBPAdm subtypes among AML, but showed divergent clinicopathologic features, mutation loci and types and differing prognostic aspects.
Collapse
Affiliation(s)
- Biao Wang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Hematology, Changzhou First People's Hospital (The Third Affiliated Hospital of Soochow University), Changzhou, China
| | - Lijun Wen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Zheng Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China; Suzhou Jsuniwell Medical Laboratory, Suzhou, China
| | - Suning Chen
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China; National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
| | - Huiying Qiu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
187
|
Sousos N, Ní Leathlobhair M, Simoglou Karali C, Louka E, Bienz N, Royston D, Clark SA, Hamblin A, Howard K, Mathews V, George B, Roy A, Psaila B, Wedge DC, Mead AJ. In utero origin of myelofibrosis presenting in adult monozygotic twins. Nat Med 2022; 28:1207-1211. [PMID: 35637336 PMCID: PMC9205768 DOI: 10.1038/s41591-022-01793-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 03/22/2022] [Indexed: 12/11/2022]
Abstract
The latency between acquisition of an initiating somatic driver mutation by a single-cell and clinical presentation with cancer is largely unknown. We describe a remarkable case of monozygotic twins presenting with CALR mutation-positive myeloproliferative neoplasms (MPNs) (aged 37 and 38 years), with a clinical phenotype of primary myelofibrosis. The CALR mutation was absent in T cells and dermal fibroblasts, confirming somatic acquisition. Whole-genome sequencing lineage tracing revealed a common clonal origin of the CALR-mutant MPN clone, which occurred in utero followed by twin-to-twin transplacental transmission and subsequent similar disease latency. Index sorting and single-colony genotyping revealed phenotypic hematopoietic stem cells (HSCs) as the likely MPN-propagating cell. Furthermore, neonatal blood spot analysis confirmed in utero origin of the JAK2V617F mutation in a patient presenting with polycythemia vera (aged 34 years). These findings provide a unique window into the prolonged evolutionary dynamics of MPNs and fitness advantage exerted by MPN-associated driver mutations in HSCs.
Collapse
Affiliation(s)
- Nikolaos Sousos
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Máire Ní Leathlobhair
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Christina Simoglou Karali
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Eleni Louka
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Nicola Bienz
- Haematology Service, Wexham Park Hospital, Frimley Health NHS Foundation Trust, Slough, UK
| | - Daniel Royston
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sally-Ann Clark
- Flow Cytometry Facility, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Angela Hamblin
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Kieran Howard
- National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Vikram Mathews
- Department of Haematology, Christian Medical College, Vellore, India
| | - Biju George
- Department of Haematology, Christian Medical College, Vellore, India
| | - Anindita Roy
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Bethan Psaila
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - David C Wedge
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK.
| | - Adam J Mead
- Medical Research Council (MRC) Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, UK.
- Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
188
|
Glück M, Dally L, Jücker M, Ehm P. JAK2-V617F is a negative regulation factor of SHIP1 protein and thus influences the AKT signaling pathway in patients with Myeloproliferative Neoplasm (MPN). Int J Biochem Cell Biol 2022; 149:106229. [PMID: 35609769 DOI: 10.1016/j.biocel.2022.106229] [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: 12/16/2021] [Revised: 04/19/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Myeloproliferative neoplasms (MPN) are a group of chronic haematological disorders. At the molecular level of MPN cells, the gain-of-function mutation V617F of the Janus kinase 2 (JAK2) leads to a constitutive activation of the downstream signaling cascade and is a conventional criteria for diagnosis. Here, the functional role of the tumor suppressor SHIP1 (SH2 domain containing inositol-5 phosphatase 1) in the pathogenesis of MPNs was investigated. METHODS Primary blood samples of MPN-patients were analysed using Western Blot technique regarding the level of SHIP1 expression. Moreover, SHIP1 and SHIP1-mutations were lentivirally transduced in the JAK2-V617F-positive UKE-1 cell line and expression was monitored over time. In addition, we examined SHIP1 reconstitution by inhibition of JAK2-V617F. Furthermore, we transfected SHIP1-expressing cells with a JAK2-V617F respectively a BCR-ABL construct and investigated changes in SHIP1 expression. RESULTS Four out of five MPN-patient samples showed a loss or a reduction in SHIP1 expression. We identified JAK2 as a negative regulator of SHIP1 expression in MPN cells and inhibition of JAK2-V617F implicates a reconstituted SHIP1 expression. This is significant because SHIP1 negatively regulates the AKT signaling pathway and in consequence the reconstitution of SHIP1 expression leads to a decreased cell growth. Moreover, we examined the impact of SHIP1 and patient-derived SHIP1-mutations on AKT phosphorylation and show the benefit of a combined therapy in MPN cells with inhibitors of the AKT/mTOR pathway. CONCLUSION In summary, the data suggest that SHIP1 may play a role during the development of MPNs and could be the basis for establishing a targeted therapy.
Collapse
Affiliation(s)
- Madeleine Glück
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Lina Dally
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Patrick Ehm
- Institute of Biochemistry and Signal Transduction, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| |
Collapse
|
189
|
Pasca S, Chifotides HT, Verstovsek S, Bose P. Mutational landscape of blast phase myeloproliferative neoplasms (MPN-BP) and antecedent MPN. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 366:83-124. [PMID: 35153007 DOI: 10.1016/bs.ircmb.2021.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Myeloproliferative neoplasms (MPN) have an inherent tendency to evolve to the blast phase (BP), characterized by ≥20% myeloblasts in the blood or bone marrow. MPN-BP portends a dismal prognosis and currently, effective treatment modalities are scarce, except for allogeneic hematopoietic stem cell transplantation (allo-HSCT) in selected patients, particularly those who achieve complete/partial remission. The mutational landscape of MPN-BP differs from de novo acute myeloid leukemia (AML) in several key aspects, such as significantly lower frequencies of FLT3 and DNMT3A mutations, and higher incidence of IDH1/2 and TP53 in MPN-BP. Herein, we comprehensively review the impact of the three signaling driver mutations (JAK2 V617F, CALR exon 9 indels, MPL W515K/L) that constitutively activate the JAK/STAT pathway, and of the other somatic non-driver mutations (epigenetic, mRNA splicing, transcriptional regulators, and mutations in signal transduction genes) that cooperatively or independently promote MPN progression and leukemic transformation. The MPN subtype, harboring two or more high-molecular risk (HMR) mutations (epigenetic regulators and mRNA splicing factors) and "triple-negative" PMF are among the critical factors that increase risk of leukemic transformation and shorten survival. Primary myelofibrosis (PMF) is the most aggressive MPN; and polycythemia vera (PV) and essential thrombocythemia (ET) are relatively indolent subtypes. In PV and ET, mutations in splicing factor genes are associated with progression to myelofibrosis (MF), and in ET, TP53 mutations predict risk for leukemic transformation. The advent of targeted next-generation sequencing and improved prognostic scoring systems for PMF inform decisions regarding allo-HSCT. The emergence of treatments targeting mutant enzymes (e.g., IDH1/2 inhibitors) or epigenetic pathways (BET and LSD1 inhibitors) along with new insights into the mechanisms of leukemogenesis will hopefully lead the way to superior management strategies and outcomes of MPN-BP patients.
Collapse
Affiliation(s)
- Sergiu Pasca
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Helen T Chifotides
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Srdan Verstovsek
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Prithviraj Bose
- Leukemia Department, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
190
|
Genetic changes during leukemic transformation to secondary acute myeloid leukemia from myeloproliferative neoplasms. Leuk Res 2022; 118:106858. [DOI: 10.1016/j.leukres.2022.106858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/30/2022] [Accepted: 05/07/2022] [Indexed: 11/22/2022]
|
191
|
Levy G, Guglielmelli P, Langmuir P, Constantinescu S. JAK inhibitors and COVID-19. J Immunother Cancer 2022; 10:jitc-2021-002838. [PMID: 35459733 PMCID: PMC9035837 DOI: 10.1136/jitc-2021-002838] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/11/2022] Open
Abstract
During SARS-CoV-2 infection, the innate immune response can be inhibited or delayed, and the subsequent persistent viral replication can induce emergency signals that may culminate in a cytokine storm contributing to the severe evolution of COVID-19. Cytokines are key regulators of the immune response and virus clearance, and, as such, are linked to the—possibly altered—response to the SARS-CoV-2. They act via a family of more than 40 transmembrane receptors that are coupled to one or several of the 4 Janus kinases (JAKs) coded by the human genome, namely JAK1, JAK2, JAK3, and TYK2. Once activated, JAKs act on pathways for either survival, proliferation, differentiation, immune regulation or, in the case of type I interferons, antiviral and antiproliferative effects. Studies of graft-versus-host and systemic rheumatic diseases indicated that JAK inhibitors (JAKi) exert immunosuppressive effects that are non-redundant with those of corticotherapy. Therefore, they hold the potential to cut-off pathological reactions in COVID-19. Significant clinical experience already exists with several JAKi in COVID-19, such as baricitinib, ruxolitinib, tofacitinib, and nezulcitinib, which were suggested by a meta-analysis (Patoulias et al.) to exert a benefit in terms of risk reduction concerning major outcomes when added to standard of care in patients with COVID-19. Yet, only baricitinib is recommended in first line for severe COVID-19 treatment by the WHO, as it is the only JAKi that has proven efficient to reduce mortality in individual randomized clinical trials (RCT), especially the Adaptive COVID-19 Treatment Trial (ACTT-2) and COV-BARRIER phase 3 trials. As for secondary effects of JAKi treatment, the main caution with baricitinib consists in the induced immunosuppression as long-term side effects should not be an issue in patients treated for COVID-19. We discuss whether a class effect of JAKi may be emerging in COVID-19 treatment, although at the moment the convincing data are for baricitinib only. Given the key role of JAK1 in both type I IFN action and signaling by cytokines involved in pathogenic effects, establishing the precise timing of treatment will be very important in future trials, along with the control of viral replication by associating antiviral molecules.
Collapse
Affiliation(s)
- Gabriel Levy
- Signal Transduction and Molecular Hematology, Ludwig Institute for Cancer Research, Brussels, Belgium.,Signal Transduction on Molecular Hematology, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium
| | - Paola Guglielmelli
- Department of Clinical and Experimental Medicine, University of Florence, Firenze, Italy.,Center of Research and Innovation for Myeloproliferative Neoplasms (CRIMM), Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Peter Langmuir
- Oncology Targeted Therapeutics, Incyte Corp, Wilmington, Delaware, USA
| | - Stefan Constantinescu
- Signal Transduction and Molecular Hematology, Ludwig Institute for Cancer Research, Brussels, Belgium .,Signal Transduction on Molecular Hematology, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Brussels, Belgium.,Nuffield Department of Medicine, Oxford University, Ludwig Institute for Cancer Research, Oxford, UK
| |
Collapse
|
192
|
Molecular Pathogenesis of Myeloproliferative Neoplasms: From Molecular Landscape to Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23094573. [PMID: 35562964 PMCID: PMC9100530 DOI: 10.3390/ijms23094573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 12/27/2022] Open
Abstract
Despite distinct clinical entities, the myeloproliferative neoplasms (MPN) share morphological similarities, propensity to thrombotic events and leukemic evolution, and a complex molecular pathogenesis. Well-known driver mutations, JAK2, MPL and CALR, determining constitutive activation of JAK-STAT signaling pathway are the hallmark of MPN pathogenesis. Recent data in MPN patients identified the presence of co-occurrence somatic mutations associated with epigenetic regulation, messenger RNA splicing, transcriptional mechanism, signal transduction, and DNA repair mechanism. The integration of genetic information within clinical setting is already improving patient management in terms of disease monitoring and prognostic information on disease progression. Even the current therapeutic approaches are limited in disease-modifying activity, the expanding insight into the genetic basis of MPN poses novel candidates for targeted therapeutic approaches. This review aims to explore the molecular landscape of MPN, providing a comprehensive overview of the role of drive mutations and additional mutations, their impact on pathogenesis as well as their prognostic value, and how they may have future implications in therapeutic management.
Collapse
|
193
|
Gagelmann N, Kröger N. Improving allogeneic stem cell transplantation in myelofibrosis. Int J Hematol 2022; 115:619-625. [PMID: 35419771 DOI: 10.1007/s12185-022-03340-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 11/30/2022]
Abstract
In this review, we will outline dimensions in which outcome of patients with myelofibrosis undergoing curative treatment can be optimized: patient selection, transplant procedure, and posttransplant prevention or treatment of relapse. For patient selection, fortunately, as with several other hematologic malignancies, the management of patients with myelofibrosis has very much entered the molecular era, with the establishment of several driver and nondriver mutations, allowing more individualized selection for treatment. For the transplant procedure itself, different conditioning intensities do not seem to play a distinctive role with regards to outcome posttransplant but still need to be compared in the molecular era. While many patients nowadays may receive ruxolitinib before transplant, recent studies may facilitate fine-tuning and integration of ruxolitinib into the transplant algorithm. The role of novel inhibitors for the transplant setting remains unclear. For the posttransplant phase, evidence remains scarce, with experiences of donor-lymphocyte infusions for relapse management but more efforts are needed in understanding relapse and identifying and treating patients at high risk for relapse.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246, Hamburg, Germany
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Martinistr 52, 20246, Hamburg, Germany.
| |
Collapse
|
194
|
Bennett C, Lawrence M, Guerrero JA, Stritt S, Waller AK, Yan Y, Mifsud RW, Ballester-Beltran J, Baig A, Mueller A, Mayer L, Warland J, Penkett CJ, Akbari P, Moreau T, Evans AL, Mookerjee S, Hoffman GJ, Saeb-Parsy K, Adams DJ, Couzens AL, Bender M, Erber WN, Nieswandt B, Read RJ, Ghevaert C. CRLF3 plays a key role in the final stage of platelet genesis and is a potential therapeutic target for thrombocythemia. Blood 2022; 139:2227-2239. [PMID: 35051265 PMCID: PMC7614665 DOI: 10.1182/blood.2021013113] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/23/2021] [Indexed: 11/20/2022] Open
Abstract
The process of platelet production has so far been understood to be a 2-stage process: megakaryocyte maturation from hematopoietic stem cells followed by proplatelet formation, with each phase regulating the peripheral blood platelet count. Proplatelet formation releases into the bloodstream beads-on-a-string preplatelets, which undergo fission into mature platelets. For the first time, we show that preplatelet maturation is a third, tightly regulated, critical process akin to cytokinesis that regulates platelet count. We show that deficiency in cytokine receptor-like factor 3 (CRLF3) in mice leads to an isolated and sustained 25% to 48% reduction in the platelet count without any effect on other blood cell lineages. We show that Crlf3-/- preplatelets have increased microtubule stability, possibly because of increased microtubule glutamylation via the interaction of CRLF3 with key members of the Hippo pathway. Using a mouse model of JAK2 V617F essential thrombocythemia, we show that a lack of CRLF3 leads to long-term lineage-specific normalization of the platelet count. We thereby postulate that targeting CRLF3 has therapeutic potential for treatment of thrombocythemia.
Collapse
Affiliation(s)
- Cavan Bennett
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
| | - Moyra Lawrence
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Jose A. Guerrero
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
| | - Simon Stritt
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Amie K. Waller
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Yahui Yan
- Cambridge Institute for Medical Research and Department of Haematology, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, England
| | - Richard W. Mifsud
- Cambridge Institute for Medical Research and Department of Haematology, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, England
| | - Jose Ballester-Beltran
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
| | - Ayesha Baig
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Annett Mueller
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Louisa Mayer
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
| | - James Warland
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Christopher J. Penkett
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
| | - Parsa Akbari
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Wort’s Causeway, Cambridge CB1 8RN, UK
- Department of Human Genetics, The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1HH, UK
| | - Thomas Moreau
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
| | - Amanda L. Evans
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Souradip Mookerjee
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Gary J. Hoffman
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, 6099, Australia
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - David J. Adams
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge, CB10 1HH, UK
| | - Amber L. Couzens
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, M5G 1X5, Canada
| | - Markus Bender
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Wendy N. Erber
- Medical School, Faculty of Health and Medical Sciences, The University of Western Australia, Crawley, WA, 6099, Australia
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, University Hospital and University of Würzburg, Josef-Schneider-Str. 2, 97080 Würzburg, Germany
| | - Randy J. Read
- Cambridge Institute for Medical Research and Department of Haematology, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge CB2 0XY, England
| | - Cedric Ghevaert
- Department of Haematology, University of Cambridge and NHS Blood and Transplant, Cambridge Blood Centre, Long Road, Cambridge CB2 0PT, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| |
Collapse
|
195
|
Tvorogov D, Thompson‐Peach CAL, Foßelteder J, Dottore M, Stomski F, Onnesha SA, Lim K, Moretti PAB, Pitson SM, Ross DM, Reinisch A, Thomas D, Lopez AF. Targeting human CALR-mutated MPN progenitors with a neoepitope-directed monoclonal antibody. EMBO Rep 2022; 23:e52904. [PMID: 35156745 PMCID: PMC8982588 DOI: 10.15252/embr.202152904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 01/02/2023] Open
Abstract
Calreticulin (CALR) is recurrently mutated in myelofibrosis via a frameshift that removes an endoplasmic reticulum retention signal, creating a neoepitope potentially targetable by immunotherapeutic approaches. We developed a specific rat monoclonal IgG2α antibody, 4D7, directed against the common sequence encoded by both insertion and deletion mutations. 4D7 selectively bound to cells co-expressing mutant CALR and thrombopoietin receptor (TpoR) and blocked JAK-STAT signalling, TPO-independent proliferation and megakaryocyte differentiation of mutant CALR myelofibrosis progenitors by disrupting the binding of CALR dimers to TpoR. Importantly, 4D7 inhibited proliferation of patient samples with both insertion and deletion CALR mutations but not JAK2 V617F and prolonged survival in xenografted bone marrow models of mutant CALR-dependent myeloproliferation. Together, our data demonstrate a novel therapeutic approach to target a problematic disease driven by a recurrent somatic mutation that would normally be considered undruggable.
Collapse
Affiliation(s)
- Denis Tvorogov
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Chloe A L Thompson‐Peach
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Johannes Foßelteder
- Department of Internal MedicineDivision of HaematologyMedical University of GrazGrazAustria
| | - Mara Dottore
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Frank Stomski
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Suraiya A Onnesha
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Kelly Lim
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Paul A B Moretti
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
| | - Stuart M Pitson
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - David M Ross
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Department of HaematologyFlinders University and Medical CentreAdelaideSAAustralia
| | - Andreas Reinisch
- Department of Internal MedicineDivision of HaematologyMedical University of GrazGrazAustria
- Department of Blood Group Serology and Transfusion MedicineMedical University of GrazGrazAustria
| | - Daniel Thomas
- Cancer ProgramPrecision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)University of AdelaideAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| | - Angel F Lopez
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Discipline of MedicineAdelaide Medical SchoolThe University of AdelaideAdelaideSAAustralia
| |
Collapse
|
196
|
Diabetes and Second Neoplasia Impact on Prognosis in Pre-Fibrotic Primary Myelofibrosis. Cancers (Basel) 2022; 14:cancers14071799. [PMID: 35406571 PMCID: PMC8997979 DOI: 10.3390/cancers14071799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary The 2016 WHO-revised classification of MPNs recognized pre-fibrotic PMF (pre-PMF) as a distinct clinical entity from both overt fibrotic PMF (overt PMF) and essential thrombocythemia (ET). In fact, while the initial presentation of pre-PMF is often an isolated thrombocytosis, thus mimicking ET, its course may be symptomatic in a non-negligible number of cases. Conversely, overt PMF patients are enriched in higher-risk categories, thus suggesting a greater propensity for disease progression than pre-PMF. Importantly, median survival is significantly reduced in overt PMF vs. pre-PMF, thereby reinforcing the appropriateness of making this distinction in clinical practice. Nevertheless, a specific prognostic model for pre-PMF is still lacking, except for thrombotic risk. The aim of the present study was therefore to identify covariates other than those commonly related to PMF, which can better define prognosis in pre-PMF patients in the real-world setting, thus resulting in more personalized and efficient therapeutic approaches. Abstract The 2016 WHO classification recognized pre-fibrotic primary myelofibrosis (pre-PMF) as a distinct entity. Nevertheless, a prognostic model specific for pre-PMF is still lacking. Our aim was to identify the most relevant clinical, histological, and driver mutation information at diagnosis to evaluate outcomes in pre-PMF patients in the real-world setting. We firstly assessed the association between IPSS or DIPSS at diagnosis and response variables in 378 pre-PMF patients. A strict association was observed between IPSS and DIPSS and occurrence of death. Other analyzed endpoints were not associated with IPSS or DIPSS as thrombo-hemorrhagic events at diagnosis or during follow-up, or did not show a clinical plausibility, as transformation into acute leukemia or overt PMF. The only covariates which were significantly associated with death were diabetes and second neoplasia, and were therefore included in two different prognostic settings: the first based on IPSS at diagnosis [class 1 vs. 0, OR (95%CIs): 3.34 (1.85–6.04); class 2 vs. 0, OR (95%CIs): 12.55 (5.04–31.24)], diabetes [OR (95%CIs): 2.95 (1.41–6.18)], and second neoplasia [OR (95%CIs): 2.88 (1.63–5.07)]; the second with DIPSS at diagnosis [class 1 vs. 0, OR (95%CIs): 3.40 (1.89–6.10); class 2 vs. 0, OR (95%CIs): 25.65 (7.62–86.42)], diabetes [OR (95%CIs): 2.89 (1.37–6.09)], and second neoplasia [OR (95%CIs): 2.97 (1.69–5.24)]. In conclusion, our study underlines the importance of other additional risk factors, such as diabetes and second neoplasia, to be evaluated, together with IPSS and DIPSS, to better define prognosis in pre-PMF patients.
Collapse
|
197
|
Enjeti AK, Agarwal R, Blombery P, Chee L, Chua CC, Grigg A, Hamad N, Iland H, Lane S, Perkins A, Singhal D, Tate C, Tiong IS, Ross DM. Panel-based gene testing in myelodysplastic/myeloproliferative neoplasm- overlap syndromes: Australasian Leukaemia and Lymphoma Group (ALLG) consensus statement. Pathology 2022; 54:389-398. [DOI: 10.1016/j.pathol.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/20/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022]
|
198
|
Loscocco GG, Vannucchi AM. Role of JAK inhibitors in myeloproliferative neoplasms: current point of view and perspectives. Int J Hematol 2022; 115:626-644. [PMID: 35352288 DOI: 10.1007/s12185-022-03335-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/06/2022] [Accepted: 03/15/2022] [Indexed: 12/29/2022]
Abstract
Classic Philadelphia-negative myeloproliferative neoplasms (MPN) include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF), classified as primary (PMF), or secondary to PV or ET. All MPN, regardless of the underlying driver mutation in JAK2/CALR/MPL, are invariably associated with dysregulation of JAK/STAT pathway. The discovery of JAK2V617F point mutation prompted the development of small molecules inhibitors of JAK tyrosine kinases (JAK inhibitors-JAKi). To date, among JAKi, ruxolitinib (RUX) and fedratinib (FEDR) are approved for intermediate and high-risk MF, and RUX is also an option for high-risk PV patients inadequately controlled by or intolerant to hydroxyurea. While not yet registered, pacritinib (PAC) and momelotinib (MMB), proved to be effective particularly in thrombocytopenic and anemic MF patients, respectively. In most cases, JAKi are effective in reducing splenomegaly and alleviating disease-related symptoms. However, almost 50% lose response by three years and dose-dependent toxicities may lead to suboptimal dosing or treatment discontinuation. To date, although not being disease-modifying agents, JAKi represent the therapeutic backbone particularly in MF patient. To optimize therapeutic strategies, many trials with drug combinations of JAKi with novel molecules are ongoing. This review critically discusses the role of JAKi in the modern management of patients with MPN.
Collapse
Affiliation(s)
- Giuseppe G Loscocco
- Department of Experimental and Clinical Medicine, University of Florence, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3 pad 27B, 50134, Florence, Italy
- Doctorate School GenOMec, University of Siena, Siena, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, University of Florence, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, Largo Brambilla, 3 pad 27B, 50134, Florence, Italy.
| |
Collapse
|
199
|
Chen R, Zhang H, Wu W, Li S, Wang Z, Dai Z, Liu Z, Zhang J, Luo P, Xia Z, Cheng Q. Antigen Presentation Machinery Signature-Derived CALR Mediates Migration, Polarization of Macrophages in Glioma and Predicts Immunotherapy Response. Front Immunol 2022; 13:833792. [PMID: 35418980 PMCID: PMC8995475 DOI: 10.3389/fimmu.2022.833792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
Immunogenicity, influenced by tumor antigenicity and antigen presenting efficiency, critically determines the effectiveness of immune checkpoint inhibitors. The role of immunogenicity has not been fully elucidated in gliomas. In this study, a large-scale bioinformatics analysis was performed to analyze the prognostic value and predictive value of antigen presentation machinery (APM) signature in gliomas. ssGSEA algorithm was used for development of APM signature and LASSO regression analysis was used for construction of APM signature-based risk score. APM signature and risk score showed favorable performance in stratifying survival and predicting tumorigenic factors of glioma patients. APM signature and risk score were also associated with different genomic features in both training cohort TCGA and validating cohort CGGA. Furthermore, APM signature-based risk score was independently validated in three external cohorts and managed to predict immunotherapy response. A prognostic nomogram was constructed based on risk score. Risk score-derived CALR was found to mediate the invasion and polarization of macrophages based on the coculture of HMC3 and U251 cells. CALR could significantly predict immunotherapy response. In conclusion, APM signature and APM signature-based risk score could help promote the clinical management of gliomas.
Collapse
Affiliation(s)
- Rui Chen
- Department of Neurosurgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wantao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Changsa, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
200
|
Analysis of factors associated with the development of myelofibrosis in polycythemia vera and essential thrombocythemia patients: a single-center experience. J Hematop 2022. [DOI: 10.1007/s12308-022-00488-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|