1
|
Najim M, Abu-Tineh M, Alshurafa A, Ibrahim MIM, Ansari S, Faraj H, Alateeg S, Akiki SJ, Yassin MA. The characteristics of CALR mutations in myeloproliferative neoplasms: a clinical experience from a tertiary care center in Qatar and a literature review. Hematology 2024; 29:2360246. [PMID: 38804886 DOI: 10.1080/16078454.2024.2360246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/21/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Myeloproliferative neoplasms (MPNs) are hematological disorders characterized by abnormal production of myeloid cells due to genetic mutations. Since 2013, researchers have identified somatic mutations in the Calreticulin (CALR) gene, primarily insertions or deletions, in two Philadelphia chromosome-negative MPNs; essential thrombocytosis (ET) and primary myelofibrosis (PMF), and occasionally in chronic myelomonocytic leukemia (CMML). This study aims to identify the various types of CALR mutations and their impact on CALR-positive MPN patients' clinical manifestations and outcomes. METHODS A single-center retrospective study was conducted. The data was collected from pre-existing records. The study was carried out on Philadelphia-negative MPN patients who were being followed up on at the NCCCR (National Center for Cancer Care and Research) to assess the clinical manifestation and outcome of disease treatment. All patients included, were followed in our center between January 1, 2008, and November 20, 2021. RESULTS A total of 50 patients with CALR-positive MPN were reviewed with a median follow-up of three years (1-11). This cohort included 31 (62%) patients with ET, 10 (20%) patients with PMF, and 9 (18%) patients with prefibrotic myelofibrosis (pre-MF). The study involved 38 (76%) male and 12 (24%) female patients. There were 16 (32%) patients diagnosed before the age of 40, 24 (48%) patients diagnosed between the ages of 40 and 60; and 10 (20%) patients diagnosed after the age of 60. Molecular analysis showed 24 (48%) patients with CALR type 1, 21 (42%) patients with CALR type 2, and 5 (10%) patients with none Type 1, none Type 2 CALR mutations. Two patients have double mutations; 1(2%) with none Type 1, none Type 2 CALR and JAK2 mutations, and 1(2%) with CALR type 1 and MPL mutations. The thrombotic events were 3 (6%) venous thromboembolisms, 3 (6%) abdominal veins thromboses, 2 (4%) strokes, and 4 (8%) ischemic cardiac events. Only 4 (8%) patients progressed to Myelofibrosis and were carrying CALR 1 mutations, and 1 (2%) patient progressed to AML with CALR 2 mutation. CONCLUSION The data shows a significant rise in CALR-positive MPN diagnoses in younger people, emphasizing the need for a better assessment tool to improve disease management and reduce complications.
Collapse
Affiliation(s)
- Mostafa Najim
- Department of Medicine, Rochester Regional Health, Unity Hospital, Rochester, NY, USA
| | - Mohammad Abu-Tineh
- Department of Medicine, Tower Health, Reading Hospital, West Reading, PA, USA
| | - Awni Alshurafa
- Department of Medical Oncology, Hematology and BMT Section, National Center for Cancer Care and Research, Doha, Qatar
| | | | - Soubiya Ansari
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Hazem Faraj
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Saif Alateeg
- Internal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Susanna Jane Akiki
- Department of Diagnostic Laboratory, Hamad Medical Corporation, Doha, Qatar
| | - Mohamed A Yassin
- Department of Medical Oncology, Hematology and BMT Section, National Center for Cancer Care and Research, Doha, Qatar
| |
Collapse
|
2
|
Sadigh S, Kim AS. Molecular Pathology of Myeloid Neoplasms: Molecular Pattern Recognition. Clin Lab Med 2024; 44:339-353. [PMID: 38821648 DOI: 10.1016/j.cll.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Despite the apparent complexity of the molecular genetic underpinnings of myeloid neoplasms, most myeloid mutational profiles can be understood within a simple framework. Somatic mutations accumulate in hematopoietic stem cells with aging and toxic insults, termed clonal hematopoiesis. These "old stem cells" mutations, predominantly in the epigenetic and RNA spliceosome pathways, act as "founding" driver mutations leading to a clonal myeloid neoplasm when sufficient in number and clone size. Subsequent mutations can create the genetic flavor of the myeloid neoplasm ("backseat" drivers) due to their enrichment in certain entities or act as progression events ("aggressive" drivers) during clonal evolution.
Collapse
Affiliation(s)
- Sam Sadigh
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Annette S Kim
- Division of Diagnostic Genetics and Genomics, Department of Pathology, University of Michigan/Michigan Medicine, 2800 Plymouth Road, NCRC 36-1221-79, Ann Arbor, MI 48109, USA.
| |
Collapse
|
3
|
Guglielmelli P, Szuber N, Gangat N, Capecchi G, Maccari C, Harnois M, Karrar O, Abdelmagid M, Balliu M, Nacca E, Atanasio A, Sestini I, Désilets A, Loscocco GG, Rotunno G, Busque L, Tefferi A, Vannucchi AM. CALR mutation burden in essential thrombocythemia and disease outcome. Blood 2024; 143:1310-1314. [PMID: 38252902 DOI: 10.1182/blood.2023023428] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/02/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
ABSTRACT Among 281 patients with essential thrombocythemia and calreticulin (CALR) mutation, we found a variant allele frequency of ≥60% to be associated with significantly shortened myelofibrosis-free survival, mostly apparent with CALR type-1 and CALR type-indeterminate mutations.
Collapse
Affiliation(s)
- Paola Guglielmelli
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Natasha Szuber
- University of Montreal, Montreal, QC, Canada
- Quebec CML-MPN Research Group, Montreal, QC, Canada
- Department of Hematology, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | | | - Giulio Capecchi
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Chiara Maccari
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | | | - Omer Karrar
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Manjola Balliu
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Elena Nacca
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Alessandro Atanasio
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Ilaria Sestini
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | | | - Giuseppe Gaetano Loscocco
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Giada Rotunno
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Lambert Busque
- University of Montreal, Montreal, QC, Canada
- Department of Hematology, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | | | - Alessandro Maria Vannucchi
- Department of Experimental and Clinical Medicine, Center of Research and Innovation of Myeloproliferative Neoplasms, Division of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| |
Collapse
|
4
|
Loscocco GG, Gesullo F, Capecchi G, Atanasio A, Maccari C, Mannelli F, Vannucchi AM, Guglielmelli P. One thousand patients with essential thrombocythemia: the Florence-CRIMM experience. Blood Cancer J 2024; 14:10. [PMID: 38238287 PMCID: PMC10796728 DOI: 10.1038/s41408-023-00968-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 01/22/2024] Open
Abstract
We describe 1000 patients with essential thrombocythemia seen at the Center Research and Innovation of Myeloproliferative Neoplasms (CRIMM), Florence, Italy, between 1980 and 2023: median age 59 years (18-95), females 65%, JAK2/CALR/MPL-mutated 66%/19%/4%, triple-negative (TN) 11%. Extreme thrombocytosis (ExT, platelets ≥1000 × 109/L) in 16%, leukocytosis (leukocytes >11 × 109/L) in 16%, and at least one cardiovascular risk factor in 52% of cases. JAK2-mutated patients were older (median 62 years) and CALR-mutated and TN (53 years for both) younger (p < 0.001). Female gender clustered with TN (76%) and JAK2 (67%) vs CALR (46%) mutations (p < 0.001). ExT clustered with CALR (type-2 more than type-1), TN and MPL, and leukocytosis with JAK2 mutation (p < 0.001). In multivariable analysis, risk factors for arterial thrombosis-free survival were age ≥60 years (HR 2.0; p < 0.001) and JAK2 mutation (HR 1.3; p = 0.02) with borderline significance for male gender (p = 0.08) and cardiovascular risk factors (p = 0.08); for venous thrombosis-free survival, JAK2 mutation (HR 1.9; p = 0.03) with borderline significance for venous thrombosis history (p = 0.07); for overall survival, older age (p < 0.001), male gender (HR 1.9; p < 0.001), absolute neutrophil count (ANC) ≥ 8 × 109/L (HR 1.8; p = 0.01), absolute lymphocyte count (ALC) < 1.7 × 109/L (HR 1.2; p = 0.03); for myelofibrosis-free survival, CALR mutation (HR 2.7; p < 0.001, particularly for CALR type 1/1-like, HR 3.3) and MPL mutation (HR 3.9; p = 0.001); for leukemia-free survival, older age (p = 0.03). Cytoreductive therapy appeared to mitigate both venous (HR 0.3; p = 0.01) and arterial thrombosis (HR 4; p = 0.04); there was a trend for aspirin in preventing arterial thrombosis recurrence. The current study provides real-world observations in essential thrombocythemia, representing a valid source document for interpreting current literature and planning future studies.
Collapse
Affiliation(s)
- Giuseppe G Loscocco
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
- Doctorate School GenOMec, University of Siena, Siena, Italy
| | - Francesca Gesullo
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Giulio Capecchi
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Alessandro Atanasio
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Chiara Maccari
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Francesco Mannelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy.
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| |
Collapse
|
5
|
Gangat N, Karrar O, Al-Kali A, Begna KH, Elliott MA, Wolanskyj-Spinner AP, Pardanani A, Hanson CA, Ketterling RP, Tefferi A. One thousand patients with essential thrombocythemia: the Mayo Clinic experience. Blood Cancer J 2024; 14:11. [PMID: 38238303 PMCID: PMC10796913 DOI: 10.1038/s41408-023-00972-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/22/2024] Open
Abstract
We describe 1000 patients with essential thrombocythemia seen at the Mayo Clinic between 1967 and 2023: median age 58 years (18-90), females 63%, JAK2/CALR/MPL-mutated 62%/27%/3%, triple-negative (TN) 8%, extreme thrombocytosis (ExT; platelets ≥1000 × 109/L) 26%, leukocytosis (leukocyte count >11 × 109/L) 20%, and abnormal karyotype 6%. JAK2-mutated patients were older (median 71 years), and CALR mutated (52 years), and TN (50 years) younger (p < 0.01). Female gender clustered with TN (73%) and JAK2 (69%) vs. CALR/MPL (49%/47%) mutations (p < 0.01). ExT clustered with CALR (type-2 more than type-1) and TN and leukocytosis with JAK2 mutation (p < 0.01). In multivariable analysis, risk factors for overall survival were older age (p < 0.01), male gender (HR 1.8), absolute neutrophil count (ANC) ≥ 8 × 109/L (HR 1.6), absolute lymphocyte count (ALC) < 1.7 × 109/L (HR 1.5), hypertension (HR 1.7), and arterial thrombosis history (HR 1.7); for leukemia-free survival, ExT (HR 2.3) and abnormal karyotype (HR 3.1); for myelofibrosis-free survival, ANC ≥ 8 × 109/L (HR 2.3) and MPL mutation (HR 3.9); for arterial thrombosis-free survival, age ≥60 years (HR 1.9), male gender (HR 1.6), arterial thrombosis history (HR 1.7), hypertension (HR 1.7), and JAK2 mutation (HR 1.8); for venous thrombosis-free survival, male gender (HR 1.8) and venous thrombosis history (HR 3.0). Associations between ExT and leukemic transformation and between ANC and fibrotic progression were limited to JAK2-mutated cases. Aspirin therapy appeared to mitigate both arterial (HR 0.4) and venous (HR 0.4) thrombosis risk. HR-based risk models delineated patients with median survivals ranging from 10 years to not reached and 20-year leukemia/myelofibrosis incidences from 3%/21% to 12.8%/49%. The current study provides both novel and confirmatory observations of essential thrombocythemia.
Collapse
Affiliation(s)
- Naseema Gangat
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Omer Karrar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Aref Al-Kali
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Curtis A Hanson
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | - Rhett P Ketterling
- Division of Laboratory Medicine and Cytogenetics, Mayo Clinic, Rochester, MN, USA
| | - Ayalew Tefferi
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
6
|
Tremblay D. Cytoreduction for ET and PV: who, what, when, and how? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:660-666. [PMID: 38066871 PMCID: PMC10727012 DOI: 10.1182/hematology.2023000451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Thrombotic complications are the primary contributor to morbidity and mortality in essential thrombocythemia (ET) and polycythemia vera (PV). Cytoreductive therapy is the main tool for primary or tertiary thrombosis prevention in these diseases. In general, high-thrombotic-risk patients and those with symptoms that may be ameliorated from cytoreductive therapy are candidates for this treatment, although the decision is highly individualized. Approved options for cytoreduction in ET and PV include hydroxyurea, long-acting interferons, anagrelide in ET, and ruxolitinib in PV. Selecting the ideal agent requires careful consideration of the toxicity profiles and individual treatment goals. In this review the existing literature on cytoreductive decisions in ET and PV is summarized, with an emphasis on risk-stratification, highlighting the need for personalized care in order to maximize the benefit of these therapies while minimizing toxicities.
Collapse
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
7
|
Gangat N, Reichard K, Tefferi A. Sodium-glucose co-transporter-2 inhibitor treatment in essential thrombocythemia: Impact on hemoglobin/hematocrit levels and outcomes among 11 consecutive patients. Am J Hematol 2023; 98:E298-E300. [PMID: 37552702 DOI: 10.1002/ajh.27048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/10/2023]
Affiliation(s)
- Naseema Gangat
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kaaren Reichard
- Division of Hematopathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ayalew Tefferi
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Mroczkowska-Bękarciak A, Wróbel T. BCR::ABL1-negative myeloproliferative neoplasms in the era of next-generation sequencing. Front Genet 2023; 14:1241912. [PMID: 37745842 PMCID: PMC10514516 DOI: 10.3389/fgene.2023.1241912] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
The classical BCR::ABL1-negative myeloproliferative neoplasms such as polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF) are clonal diseases with the presence of characteristic "driver mutations" in one of the genes: JAK2, CALR, or MPL. The search for mutations in these three genes is required for the diagnosis of MPNs. Nevertheless, the progress that has been made in the field of molecular genetics has opened a new era in medicine. The search for additional mutations in MPNs is helpful in assessing the risk stratification, disease progression, transformation to acute myeloid leukemia (AML), or choosing the right treatment. In some cases, advanced technologies are needed to find a clonal marker of the disease and establish a diagnosis. This review focuses on how the use of new technologies like next-generation sequencing (NGS) helps in the diagnosis of BCR::ABL1-negative myeloproliferative neoplasms.
Collapse
|
9
|
Iurlo A, Bucelli C, Cattaneo D. Essential Thrombocythemia in Adolescents and Young Adults: Clinical Aspects, Treatment Options and Unmet Medical Needs. Curr Treat Options Oncol 2023; 24:802-820. [PMID: 37195587 DOI: 10.1007/s11864-023-01099-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2023] [Indexed: 05/18/2023]
Abstract
OPINION STATEMENT Current treatment of essential thrombocythemia (ET) should primarily prevent thrombo-hemorrhagic events, without increasing the rate of fibrotic progression or leukemic evolution, and secondarily control microvascular symptoms. Unlike other classic BCR::ABL1-negative myeloproliferative neoplasms, ET is frequently diagnosed in adolescents and young adults (AYA), defined as individuals aged 15 to 39 years, in up to 20% of patients. However, since the current risk stratification of this disease is based on models, including that of ELN, IPSET-Thrombosis and its revised version, mainly applied to an older patients' population, international guidelines are needed that specifically consider how to evaluate the prognosis of AYAs with ET. Furthermore, although ET is the most frequent MPN among AYA subjects, there is a lack of specific recommendations on how to treat it in this subgroup of patients, as management decisions are typically extrapolated from those for the elderly. Accordingly, since AYAs with ET represent a unique disease subset defined by attenuated genetic risk, more indolent phenotype, and longer survival than their older counterparts, treatment selection requires special attention to specific issues such as the risk of fibrotic/leukemic transformation, carcinogenicity, and fertility. This review article will provide a comprehensive overview of the diagnosis, prognostic stratification, and possible therapeutic approaches for AYA patients with ET, including antiplatelets/anticoagulants and cytoreductive agents, with a focus on pregnancy management in real-life clinical practice.
Collapse
Affiliation(s)
- Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy.
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
10
|
Andrews C, Conneally E, Langabeer SE. Molecular diagnostic criteria of myeloproliferative neoplasms. Expert Rev Mol Diagn 2023; 23:1077-1090. [PMID: 37999991 DOI: 10.1080/14737159.2023.2277370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/26/2023] [Indexed: 11/26/2023]
Abstract
INTRODUCTION Myeloproliferative neoplasms (MPN) are a heterogeneous group of clonal hematopoietic stem cell neoplasms characterized by the driver mutations JAK2, CALR, and MPL. These mutations cause constitutive activation of JAK-STAT signaling, which is central to pathogenesis of MPNs. Next-generation sequencing has further expanded the molecular landscape allowing for improved diagnostics, prognostication, and targeted therapy. AREAS COVERED This review aims to address current understanding of the molecular diagnosis of MPN not only through improved awareness of the driver mutations but also the disease modifying mutations. In addition, other genetic factors such as clonal hematopoiesis of indeterminate potential (CHIP), order of mutation, and mutation co-occurrence are discussed and how these factors influence disease initiation and ultimately progression. How this molecular information is incorporated into risk stratification models allowing for earlier intervention and targeted therapy in the future will be addressed further. EXPERT OPINION The genomic landscape of the MPN has evolved in the last 15 years with integration of next-generation sequencing becoming the gold standard of MPN management. Although diagnostics and prognostication have become more personalized, additional studies are required to translate these molecular findings into targeted therapy therefore improving patient outcomes.
Collapse
Affiliation(s)
- Claire Andrews
- Department of Haematology, St. Vincent's University Hospital, Dublin, Ireland
| | | | | |
Collapse
|
11
|
Giraldo-Rincón AI, Naranjo Molina S, Gomez-Lopera N, Aguirre Acevedo D, Ucroz Benavidez A, Gálvez Cárdenas K, Cuellar Ambrosí F, Torres JD, Ospina S, Palacio K, Gaviria Jaramillo L, Muñeton CM, Vasquez Palacio G. JAK2, CALR, and MPL Mutation Profiles in Colombian patients with BCR-ABL Negative Myeloproliferative Neoplasms. Colomb Med (Cali) 2023; 54:e2035353. [PMID: 38111518 PMCID: PMC10726695 DOI: 10.25100/cm.v54i3.5353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 04/25/2023] [Indexed: 12/20/2023] Open
Abstract
Background Among the chronic myeloproliferative neoplasms (MPNs) not associated with BCR-ABL mutations are polycythemia vera, primary myelofibrosis, and essential thrombocythemia. These diseases are caused by mutations in genes, such as the JAK2, MPL, and CALR genes, which participate in regulating the JAK-STAT signaling pathway. Objective This study aimed to establish the frequencies of mutations in the JAK2, MPL, and CALR genes in a group of Colombian patients with a negative clinical diagnosis of BCR-ABL chronic myeloproliferative neoplasms. Methods The JAK2 V617F and MPL W515K mutations and deletions or insertions in exon 9 of the CALR gene were analyzed in 52 Colombian patients with polycythemia vera, primary myelofibrosis, and essential thrombocythemia. Results The JAK2V617F mutation was carried by 51.9% of the patients, the CALR mutation by 23%, and the MPL mutation by 3.8%; 23% were triple-negative for the mutations analyzed. In these neoplasms, 6 mutation types in CALR were identified, one of which has not been previously reported. Additionally, one patient presented a double mutation in both the CALR and JAK2 genes. Regarding the hematological results for the mutations, significant differences were found in the hemoglobin level, hematocrit level, and platelet count among the three neoplasms. Conclusion Thus, this study demonstrates the importance of the molecular characterization of the JAK2, CALR and MPL mutations in Colombian patients (the genetic context of which remains unclear in the abovementioned neoplasms) to achieve an accurate diagnosis, a good prognosis, adequate management, and patient survival.
Collapse
Affiliation(s)
| | - Sara Naranjo Molina
- Universidad de Antioquia, Facultad de Medicina, Unidad de Genética Médica, Medellín, Colombia
| | - Natalia Gomez-Lopera
- Universidad de Antioquia, Facultad de Medicina, Unidad de Genética Médica, Medellín, Colombia
| | | | - Andrea Ucroz Benavidez
- Universidad de Antioquia, Facultad de Medicina, Unidad de Genética Médica, Medellín, Colombia
| | | | | | | | - Sigifredo Ospina
- Hospital Universitario San Vicente Fundación, Medellín, Colombia
| | - Katherine Palacio
- Universidad de Antioquia, Facultad de Medicina, Unidad de Genética Médica, Medellín, Colombia
| | | | - Carlos Mario Muñeton
- Universidad de Antioquia, Facultad de Medicina, Unidad de Genética Médica, Medellín, Colombia
| | - Gonzalo Vasquez Palacio
- Universidad de Antioquia, Facultad de Medicina, Unidad de Genética Médica, Medellín, Colombia
| |
Collapse
|
12
|
Wang YH, Chen YJ, Lai YH, Wang MC, Chen YY, Wu YY, Yang YR, Tsou HY, Li CP, Hsu CC, Huang CE, Chen CC. Mutation-Driven S100A8 Overexpression Confers Aberrant Phenotypes in Type 1 CALR-Mutated MPN. Int J Mol Sci 2023; 24:8747. [PMID: 37240094 PMCID: PMC10217897 DOI: 10.3390/ijms24108747] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Numerous pathogenic CALR exon 9 mutations have been identified in myeloproliferative neoplasms (MPN), with type 1 (52bp deletion; CALRDEL) and type 2 (5bp insertion; CALRINS) being the most prevalent. Despite the universal pathobiology of MPN driven by various CALR mutants, it is unclear why different CALR mutations result in diverse clinical phenotypes. Through RNA sequencing followed by validation at the protein and mRNA levels, we found that S100A8 was specifically enriched in CALRDEL but not in CALRINS MPN-model cells. The expression of S100a8 could be regulated by STAT3 based on luciferase reporter assay complemented with inhibitor treatment. Pyrosequencing demonstrated relative hypomethylation in two CpG sites within the potential pSTAT3-targeting S100a8 promoter region in CALRDEL cells as compared to CALRINS cells, suggesting that distinct epigenetic alteration could factor into the divergent S100A8 levels in these cells. The functional analysis confirmed that S100A8 non-redundantly contributed to accelerated cellular proliferation and reduced apoptosis in CALRDEL cells. Clinical validation showed significantly enhanced S100A8 expression in CALRDEL-mutated MPN patients compared to CALRINS-mutated cases, and thrombocytosis was less prominent in those with S100A8 upregulation. This study provides indispensable insights into how different CALR mutations discrepantly drive the expression of specific genes that contributes to unique phenotypes in MPN.
Collapse
Affiliation(s)
- Ying-Hsuan Wang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Ying-Ju Chen
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Yi-Hua Lai
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Ming-Chung Wang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Yi-Yang Chen
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Yu-Ying Wu
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Yao-Ren Yang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Hsing-Yi Tsou
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Chian-Pei Li
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Chia-Chen Hsu
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
| | - Cih-En Huang
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chih-Cheng Chen
- Division of Hematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan; (Y.-H.W.); (Y.-J.C.); (Y.-H.L.); (Y.-Y.C.); (Y.-Y.W.); (Y.-R.Y.); (H.-Y.T.); (C.-P.L.); (C.-C.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
13
|
Desikan H, Kaur A, Pogozheva ID, Raghavan M. Effects of calreticulin mutations on cell transformation and immunity. J Cell Mol Med 2023; 27:1032-1044. [PMID: 36916035 PMCID: PMC10098294 DOI: 10.1111/jcmm.17713] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are cancers involving dysregulated production and function of myeloid lineage hematopoietic cells. Among MPNs, Essential thrombocythemia (ET), Polycythemia Vera (PV) and Myelofibrosis (MF), are driven by mutations that activate the JAK-STAT signalling pathway. Somatic mutations of calreticulin (CRT), an endoplasmic reticulum (ER)-localized lectin chaperone, are driver mutations in approximately 25% of ET and 35% of MF patients. The MPN-linked mutant CRT proteins have novel frameshifted carboxy-domain sequences and lack an ER retention motif, resulting in their secretion. Wild type CRT is a regulator of ER calcium homeostasis and plays a key role in the assembly of major histocompatibility complex (MHC) class I molecules, which are the ligands for antigen receptors of CD8+ T cells. Mutant CRT-linked oncogenesis results from the dysregulation of calcium signalling in cells and the formation of stable complexes of mutant CRT with myeloproliferative leukemia (MPL) protein, followed by downstream activation of the JAK-STAT signalling pathway. The intricate participation of CRT in ER protein folding, calcium homeostasis and immunity suggests the involvement of multiple mechanisms of mutant CRT-linked oncogenesis. In this review, we highlight recent findings related to the role of MPN-linked CRT mutations in the dysregulation of calcium homeostasis, MPL activation and immunity.
Collapse
Affiliation(s)
- Harini Desikan
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Amanpreet Kaur
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Irina D. Pogozheva
- Department of Medicinal ChemistryCollege of Pharmacy, University of MichiganAnn ArborMichiganUSA
| | - Malini Raghavan
- Department of Microbiology and ImmunologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
14
|
Abbou N, Piazzola P, Gabert J, Ernest V, Arcani R, Couderc AL, Tichadou A, Roche P, Farnault L, Colle J, Ouafik L, Morange P, Costello R, Venton G. Impact of Molecular Biology in Diagnosis, Prognosis, and Therapeutic Management of BCR::ABL1-Negative Myeloproliferative Neoplasm. Cells 2022; 12:cells12010105. [PMID: 36611899 PMCID: PMC9818322 DOI: 10.3390/cells12010105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
BCR::ABL1-negative myeloproliferative neoplasms (MPNs) include three major subgroups-polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF)-which are characterized by aberrant hematopoietic proliferation with an increased risk of leukemic transformation. Besides the driver mutations, which are JAK2, CALR, and MPL, more than twenty additional mutations have been identified through the use of next-generation sequencing (NGS), which can be involved with pathways that regulate epigenetic modifications, RNA splicing, or DNA repair. The aim of this short review is to highlight the impact of molecular biology on the diagnosis, prognosis, and therapeutic management of patients with PV, ET, and PMF.
Collapse
Affiliation(s)
- Norman Abbou
- Molecular Biology Laboratory, North University Hospital, 13015 Marseille, France
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
| | - Pauline Piazzola
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - Jean Gabert
- Molecular Biology Laboratory, North University Hospital, 13015 Marseille, France
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
| | - Vincent Ernest
- Hematology Laboratory, Timone University Hospital, 13005 Marseille, France
| | - Robin Arcani
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
- Department of Internal Medicine, Timone University Hospital, 13005 Marseille, France
| | - Anne-Laure Couderc
- Department of Geriatrics, South University Hospital, 13005 Marseille, France
| | - Antoine Tichadou
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - Pauline Roche
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - Laure Farnault
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - Julien Colle
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
| | - L’houcine Ouafik
- CNRS, INP, Institute of Neurophysiopathol, Aix-Marseille Université, 13005 Marseille, France
- APHM, CHU Nord, Service d’Onco-Biologie, Aix-Marseille Université, 13005 Marseille, France
| | - Pierre Morange
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
- Hematology Laboratory, Timone University Hospital, 13005 Marseille, France
| | - Régis Costello
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
- TAGC, INSERM, UMR1090, Aix-Marseille University, 13005 Marseille, France
| | - Geoffroy Venton
- INSERM, INRAE, C2VN, Aix-Marseille University, 13005 Marseille, France
- Hematology and Cellular Therapy Department, Conception University Hospital, 13005 Marseille, France
- TAGC, INSERM, UMR1090, Aix-Marseille University, 13005 Marseille, France
- Correspondence: ; Tel.: +33-4-91-38-41-52
| |
Collapse
|
15
|
Larsen MK, Skov V, Kjær L, Møller‐Palacino NA, Pedersen RK, Andersen M, Ottesen JT, Cordua S, Poulsen HE, Dahl M, Knudsen TA, Eickhardt‐Dalbøge CS, Koschmieder S, Pedersen KM, Çolak Y, Bojesen SE, Nordestgaard BG, Stiehl T, Hasselbalch HC, Ellervik C. Clonal haematopoiesis of indeterminate potential and impaired kidney function-A Danish general population study with 11 years follow-up. Eur J Haematol 2022; 109:576-585. [PMID: 36054308 PMCID: PMC9804367 DOI: 10.1111/ejh.13845] [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: 05/26/2022] [Accepted: 08/08/2022] [Indexed: 01/05/2023]
Abstract
The myeloproliferative neoplasms are associated with chronic kidney disease but whether clonal haematopoiesis of indeterminate potential (CHIP) is associated with impaired kidney function is unknown. In the Danish General Suburban Population Study (N = 19 958) from 2010 to 2013, 645 individuals were positive for JAK2V617F (N = 613) or CALR (N = 32) mutations. Mutation-positive individuals without haematological malignancy were defined as having CHIP (N = 629). We used multiple and inverse probability weighted (IPW)-adjusted linear regression analysis to estimate adjusted mean (95% confidence interval) differences in estimated glomerular filtration rate (eGFR; ml/min/1.73 m2 ) by mutation status, variant allele frequency (VAF%), blood cell counts, and neutrophil-to-lymphocyte ratio (NLR). We performed 11-year longitudinal follow-up of eGFR in all individuals. Compared to CHIP-negative individuals, the mean differences in eGFR were -5.6 (-10.3, -0.8, p = .02) for CALR, -11.9 (-21.4, -2.4, p = 0.01) for CALR type 2, and -10.1 (-18.1, -2.2, p = .01) for CALR with VAF ≥ 1%. The IPW-adjusted linear regression analyses showed similar results. NLR was negatively associated with eGFR. Individuals with CALR type 2 had a worse 11-year longitudinal follow-up on eGFR compared to CHIP-negative individuals (p = .004). In conclusion, individuals with CALR mutations, especially CALR type 2, had impaired kidney function compared to CHIP-negative individuals as measured by a lower eGFR at baseline and during 11-year follow-up.
Collapse
Affiliation(s)
- Morten K. Larsen
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Vibe Skov
- Department of HaematologyZealand University HospitalRoskildeDenmark
| | - Lasse Kjær
- Department of HaematologyZealand University HospitalRoskildeDenmark
| | | | | | - Morten Andersen
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark
| | - Johnny T. Ottesen
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark
| | - Sabrina Cordua
- Department of HaematologyCopenhagen University Hospital RigshospitaletCopenhagenDenmark
| | - Henrik E. Poulsen
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of EndocrinologyCopenhagen University Hospital, Bispebjerg Frederiksberg HospitalCopenhagenDenmark,Department of CardiologyCopenhagen University Hospital, Nordsjællands HospitalHillerødDenmark
| | - Morten Dahl
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical BiochemistryZealand University HospitalKøgeDenmark
| | - Trine A. Knudsen
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Christina Schjellerup Eickhardt‐Dalbøge
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Steffen Koschmieder
- Department of Haematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of MedicineRWTH Aachen UniversityAachenGermany
| | - Kasper M. Pedersen
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark
| | - Yunus Çolak
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark,Department of Respiratory MedicineCopenhagen University Hospital, Herlev and Gentofte HospitalDenmark
| | - Stig E. Bojesen
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark
| | - Børge G. Nordestgaard
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Clinical Biochemistry and the Copenhagen General Population StudyCopenhagen University Hospital, Herlev and Gentofte HospitalHerlevDenmark
| | - Thomas Stiehl
- Department of Science and EnvironmentRoskilde UniversityRoskildeDenmark,Institute for Computational Biomedicine ‐ Disease ModellingFaculty of Medicine, RWTH Aachen UniversityAachenGermany
| | - Hans C. Hasselbalch
- Department of HaematologyZealand University HospitalRoskildeDenmark,Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Christina Ellervik
- Department Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark,Department of Data SupportRegion ZealandDenmark,Department of PathologyHarvard Medical SchoolBostonUSA
| |
Collapse
|
16
|
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: 7.5] [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
|
17
|
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: 16] [Impact Index Per Article: 8.0] [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
|
18
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [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
|
19
|
CALR type 1 mutations are associated with an increased incidence of myelofibrosis in young male patients. Ir J Med Sci 2022; 192:591-593. [PMID: 35672563 PMCID: PMC10066159 DOI: 10.1007/s11845-022-03047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/02/2022] [Indexed: 10/18/2022]
Abstract
Abstract
Background
Calreticulin (CALR) mutations are commonly identified in patients with essential thrombocythaemia or myelofibrosis. CALR type 1 mutations are known to have a higher overall incidence in males but little is known about the risks of mutation subtypes on myelofibrotic change across patient age and sex.
Aims
To identify differences in the incidence of myelofibrotic change within subgroups of patients with CALR type 1 mutations.
Methods
All patients with a positive CALR exon 9 mutation identified within our unit between February 2016 and September 2020 were reviewed with note taken of patient sex, age at diagnosis, initial MPN diagnosis, and subsequent disease transformation.
Results
In our cohort, young male patients with CALR type 1 mutations were shown to be at significantly increased risk of myelofibrosis compared to age matched female patients.
Conclusions
Male patients have a worse myeloproliferative neoplasm phenotype than female patients with it occurring at a younger age and being more myelofibrotic in nature. Further investigation is needed into the reasons for this variability.
Collapse
|
20
|
Jain T, Tsai HL, DeZern AE, Gondek LP, Elmariah H, Bolaños-Meade J, Luznik L, Fuchs E, Ambinder R, Gladstone DE, Imus P, Webster J, Prince G, Ghiaur G, Smith BD, Ali SA, Ambinder A, Dalton WB, Gocke CB, Huff CA, Gojo I, Swinnen L, Wagner-Johnston N, Borrello I, Varadhan R, Levis M, Jones RJ. Post-Transplantation Cyclophosphamide-Based Graft- versus-Host Disease Prophylaxis with Nonmyeloablative Conditioning for Blood or Marrow Transplantation for Myelofibrosis. Transplant Cell Ther 2022; 28:259.e1-259.e11. [PMID: 35158092 PMCID: PMC9081210 DOI: 10.1016/j.jtct.2022.02.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/20/2022]
Abstract
We describe outcomes after post-transplantation cyclophosphamide and nonmyeloablative conditioning-based allogeneic blood or marrow transplantation for myelofibrosis using matched or mismatched related or unrelated donors. The conditioning regimen consisted of fludarabine, cyclophosphamide, and total body irradiation. Forty-two patients were included, with a median age of 63 years, of whom 19% had Dynamic International Prognostic Scoring System (DIPSS)-plus intermediate-1 risk, 60% had intermediate-2 risk, and 21% had high-risk disease, and 60% had at least 1 high-risk somatic mutation. More than 90% of patients engrafted neutrophils, at a median of 19.5 days, and 7% experienced graft failure. At 1 year and 3 years, respectively, overall survival was 65% and 60%, relapse-free survival was 65% and 31%, relapse was 5% and 40%, and nonrelapse mortality was 30% and 30%. Acute graft-versus-host disease grade 3-4 was seen in 17% of patients at 1 year, and chronic graft-versus-host disease requiring systemic therapy in occurred in 12% patients. Spleen size ≥17 cm or prior splenectomy was associated with inferior relapse-free survival (hazard ratio [HR], 3.50; 95% confidence interval [CI], 1.18 to 10.37; P = .02) and higher relapse rate (subdistribution HR [SDHR] not calculable; P = .01). Age >60 years (SDHR, 0.26; 95% CI, 0.08 to 0.80, P = .02) and receipt of peripheral blood grafts (SDHR, 0.34; 95% CI, 0.11 to 0.99; P = .05) were associated with a lower risk of relapse. In our limited sample, the presence of a high-risk mutation was not statistically significantly associated with an inferior outcome, although ASXL1 was suggestive of inferior survival (SDHR, 2.36; 95% CI, 0.85 to 6.6; P = .09). Overall, this approach shows outcomes comparable those of to previously reported approaches and underscores the importance of spleen size in the evaluation of transplantation candidates.
Collapse
Affiliation(s)
- Tania Jain
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Hua-Ling Tsai
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Amy E DeZern
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lukasz P Gondek
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hany Elmariah
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida
| | - Javier Bolaños-Meade
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Leonido Luznik
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ephraim Fuchs
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Douglas E Gladstone
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Philip Imus
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan Webster
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gabrielle Prince
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gabriel Ghiaur
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - B Douglas Smith
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Syed Abbas Ali
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander Ambinder
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William B Dalton
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christian B Gocke
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carol Ann Huff
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ivana Gojo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lode Swinnen
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nina Wagner-Johnston
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ivan Borrello
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Johns Hopkins/Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Mark Levis
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Richard J Jones
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
21
|
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: 2.0] [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
|
22
|
Tefferi A, Gangat N, Pardanani A, Crispino JD. Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape. Cancer Res 2022; 82:749-763. [PMID: 34911786 PMCID: PMC9306313 DOI: 10.1158/0008-5472.can-21-2930] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/18/2021] [Accepted: 11/30/2021] [Indexed: 01/07/2023]
Abstract
Primary myelofibrosis (PMF) is one of three myeloproliferative neoplasms (MPN) that are morphologically and molecularly inter-related, the other two being polycythemia vera (PV) and essential thrombocythemia (ET). MPNs are characterized by JAK-STAT-activating JAK2, CALR, or MPL mutations that give rise to stem cell-derived clonal myeloproliferation, which is prone to leukemic and, in case of PV and ET, fibrotic transformation. Abnormal megakaryocyte proliferation is accompanied by bone marrow fibrosis and characterizes PMF, while the clinical phenotype is pathogenetically linked to ineffective hematopoiesis and aberrant cytokine expression. Among MPN-associated driver mutations, type 1-like CALR mutation has been associated with favorable prognosis in PMF, while ASXL1, SRSF2, U2AF1-Q157, EZH2, CBL, and K/NRAS mutations have been shown to be prognostically detrimental. Such information has enabled development of exclusively genetic (GIPSS) and clinically integrated (MIPSSv2) prognostic models that facilitate individualized treatment decisions. Allogeneic stem cell transplantation remains the only treatment modality in MF with the potential to prolong survival, whereas drug therapy, including JAK2 inhibitors, is directed mostly at the inflammatory component of the disease and is therefore palliative in nature. Similarly, disease-modifying activity remains elusive for currently available investigational drugs, while their additional value in symptom management awaits controlled confirmation. There is a need for genetic characterization of clinical observations followed by in vitro and in vivo preclinical studies that will hopefully identify therapies that target the malignant clone in MF to improve patient outcomes.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.,Corresponding Author: Ayalew Tefferi, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905. Phone: 507-284-2511; Fax: 507-266-4972; E-mail:
| | - Naseema Gangat
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - John D. Crispino
- Division of Experimental Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
23
|
Fabris S, Cattaneo D, Salerio S, Bucelli C, Ciceri G, Pasquale R, Todoerti K, Gianelli U, Baldini L, Neri A, Iurlo A. Impact on thrombotic risk of canonical and atypical CALR mutations in essential thrombocythemia. A single-center cohort study. Thromb Res 2022; 210:67-69. [PMID: 35016075 DOI: 10.1016/j.thromres.2021.12.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/05/2023]
Affiliation(s)
- Sonia Fabris
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Simone Salerio
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Ciceri
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Raffaella Pasquale
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Katia Todoerti
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Umberto Gianelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy; Division of Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Baldini
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Antonino Neri
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
24
|
Essential Thrombocythemia in Children and Adolescents. Cancers (Basel) 2021; 13:cancers13236147. [PMID: 34885256 PMCID: PMC8656963 DOI: 10.3390/cancers13236147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary Among chronic Ph-negative myeloproliferative neoplasms, essential thrombocythemia is found in children with low but increasing incidence. The diagnostic and clinical features do not completely overlap with ET of adult age. A significant number of cases, in fact, do not meet the criteria of clonality, and many cases require extensive clinical evaluation to exclude secondary, reactive forms. Therefore, histological analysis of bone marrow biopsy is necessary, and its use should be enforced. The clinical course appears to be more benign, at least within the first decades of observation, with the incidence of thrombotic events being much lower than in adults (4 % vs. 30%). Hemorrhages are mostly irrelevant. Therefore, the management should be carefully adapted to the individual patient, balancing the risk of future complications with long-term collateral effects of any drug. This review analyzes the peculiarities of the disease facing similarities and differences with adult scenarios. Abstract This paper reviews the features of pediatric essential thrombocythemia (ET). ET is a rare disease in children, challenging pediatric and adult hematologists alike. The current WHO classification acknowledges classical Philadelphia-negative MPNs and defines diagnostic criteria, mainly encompassing adult cases. The presence of one of three driver mutations (JAK2V617F, CALR, and MPL mutations) represent the proof of clonality typical of ET. Pediatric ET cases are thus usually confronted by adult approaches. These can fit only some patients, because only 25–40% of cases present one of the driver mutations. The diagnosis of hereditary, familial thrombocytosis and the exclusion of reactive/secondary thrombocytosis must be part of the diagnostic process in children and can clarify most of the negative cases. Still, many children present a clinical, histological picture of ET, with a molecular triple wild-type status. Moreover, prognosis seems more benign, at least within the first few decades of follow-up. Thrombotic events are rare, and only minor hemorrhages are ordinarily observed. As per the management, the need to control symptoms must be balanced with the collateral effects of lifelong drug therapy. We conclude that these differences concert a compelling case for a very careful therapeutic approach and advocate for the importance of further cooperative studies.
Collapse
|
25
|
Coltro G, Loscocco GG, Vannucchi AM. Classical Philadelphia-negative myeloproliferative neoplasms (MPNs): A continuum of different disease entities. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:1-69. [PMID: 34756241 DOI: 10.1016/bs.ircmb.2021.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Classical Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell-derived disorders characterized by uncontrolled proliferation of differentiated myeloid cells and close pathobiologic and clinical features. According to the 2016 World Health Organization (WHO) classification, MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The 2016 revision aimed in particular at strengthening the distinction between masked PV and JAK2-mutated ET, and between prefibrotic/early (pre-PMF) and overt PMF. Clinical manifestations in MPNs include constitutional symptoms, microvascular disorders, thrombosis and bleeding, splenomegaly secondary to extramedullary hematopoiesis, cytopenia-related symptoms, and progression to overt MF and acute leukemia. A dysregulation of the JAK/STAT pathway is the unifying mechanistic hallmark of MPNs, and is guided by somatic mutations in driver genes including JAK2, CALR and MPL. Additional mutations in myeloid neoplasm-associated genes have been also identified, with established prognostic relevance, particularly in PMF. Prognostication of MPN patients relies on disease-specific clinical models. The increasing knowledge of MPN biology led to the development of integrated clinical and molecular prognostic scores that allow a more refined stratification. Recently, the therapeutic landscape of MPNs has been revolutionized by the introduction of potent, selective JAK inhibitors (ruxolitinib, fedratinib), that proved effective in controlling disease-related symptoms and splenomegaly, yet leaving unmet critical needs, owing the lack of disease-modifying activity. In this review, we will deal with molecular, clinical, and therapeutic aspects of the three classical MPNs aiming at highlighting either shared characteristics, that overall define a continuum within a single disease family, and uniqueness, at the same time.
Collapse
Affiliation(s)
- Giacomo Coltro
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giuseppe G Loscocco
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro M Vannucchi
- CRIMM, Center for Research and Innovation of Myeloproliferative Neoplasms, AOU Careggi, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
26
|
Sadigh S, Kim AS. Molecular Pathology of Myeloid Neoplasms: Molecular Pattern Recognition. Surg Pathol Clin 2021; 14:517-528. [PMID: 34373100 DOI: 10.1016/j.path.2021.05.013] [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: 10/20/2022]
Abstract
Despite the apparent complexity of the molecular genetic underpinnings of myeloid neoplasms, most myeloid mutational profiles can be understood within a simple framework. Somatic mutations accumulate in hematopoietic stem cells with aging and toxic insults, termed clonal hematopoiesis. These "old stem cells" mutations, predominantly in the epigenetic and RNA spliceosome pathways, act as "founding" driver mutations leading to a clonal myeloid neoplasm when sufficient in number and clone size. Subsequent mutations can create the genetic flavor of the myeloid neoplasm ("backseat" drivers) due to their enrichment in certain entities or act as progression events ("aggressive" drivers) during clonal evolution.
Collapse
Affiliation(s)
- Sam Sadigh
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA
| | - Annette S Kim
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Yang E, Wang M, Wang Z, Li Y, Wang X, Ming J, Xiao H, Quan R, Liu W, Hu X. Comparison of the effects between MPL and JAK2V617F on thrombosis and peripheral blood cell counts in patients with essential thrombocythemia: a meta-analysis. Ann Hematol 2021; 100:2699-2706. [PMID: 34383101 PMCID: PMC8510937 DOI: 10.1007/s00277-021-04617-6] [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: 04/17/2021] [Accepted: 07/16/2021] [Indexed: 01/03/2023]
Abstract
To assess the effects between MPL and JAK2V617F on the thrombosis risk and peripheral blood cell counts in patients with essential thrombocythemia (ET), we identified eligible studies from PubMed, Embase, and the Cochrane Library. Seven studies were ultimately included in this meta-analysis. All studies reported the peripheral blood cell counts of ET patients, and three of them reported the eligible thrombotic events. In comparing the effect of MPL versus JAK2V617F on thrombosis, 1257 ET patients (73 MPL + and 1184 JAK2V617F +) were included. MPL-positive (MPL +) ET patients had a higher risk of thrombosis than JAK2V617F-positive (JAK2V617F +) ET patients [RR = 1.80 (1.08-3.01), P = 0.025]. And 3453 ET patients (138 MPL + and 3315 JAK2V617F +) were included in the comparison of peripheral blood cell counts. Platelet counts of MPL + ET patients were higher than that of JAK2V617F + ET patients [WMD = 81.18 (31.77-130.60), P = 0.001]. MPL + ET patients had lower hemoglobin [WMD = - 11.66 (- 14.32 to - 9.00), P = 0.000] and white blood cell counts [WMD = - 1.01 (- 1.47 to - 0.56), P = 0.000] than JAK2V617F + ET patients. These findings indicate that the MPL mutation is a high-risk factor for thrombosis in ET patients, and it may be rational to include MPL mutation in the revised IPSET as a criterion for thrombosis prediction scores. And given the differences in peripheral blood, it is necessary to further study whether MPL + ET patients differ from JAK2V617F + ET patients in bleeding and survival.
Collapse
Affiliation(s)
- Erpeng Yang
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.,Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Mingjing Wang
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.,Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ziqing Wang
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.,Xiyuan Clinical Medical College of Beijing University of Traditional Chinese Medicine, Beijing, 100029, China
| | - Yujin Li
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.,Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xueying Wang
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.,Xiyuan Clinical Medical College of Beijing University of Traditional Chinese Medicine, Beijing, 100029, China
| | - Jing Ming
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Haiyan Xiao
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Richeng Quan
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China
| | - Weiyi Liu
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.
| | - Xiaomei Hu
- Department of Haematology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, No.1 Xiyuan Caochang Road, Haidian District, Beijing, 100091, China.
| |
Collapse
|
28
|
The Prognostic Role of Cytogenetics Analysis in Philadelphia Negative Myeloproliferative Neoplasms. ACTA ACUST UNITED AC 2021; 57:medicina57080813. [PMID: 34441019 PMCID: PMC8398709 DOI: 10.3390/medicina57080813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 11/16/2022]
Abstract
Myeloproliferative neoplasms (MPNs) are clonal stem cell disorders characterized collectively by clonal proliferation of myeloid cells with variable morphologic maturity and hematopoietic efficiency. Although the natural history of these neoplasms can be measured sometimes in decades more than years, the cytogenetics analysis can offer useful information regarding the prognosis. Cytogenetics has a well-established prognostic role in acute leukemias and in myelodysplastic syndromes, where it drives the clinical decisions. NGS techniques can find adverse mutations with clear prognostic value and are currently included in the prognostic evaluation of MPNs in scores such as MIPSS, GIPSS, MIPSS-PV, and MIPSS-ET. We suggest that cytogenetics (considering its availability and relative cost) has a role regarding prognostic and therapeutic decisions.
Collapse
|
29
|
Integration of Molecular Information in Risk Assessment of Patients with Myeloproliferative Neoplasms. Cells 2021; 10:cells10081962. [PMID: 34440731 PMCID: PMC8391705 DOI: 10.3390/cells10081962] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/30/2022] Open
Abstract
Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) are clonal disorders of a hematopoietic stem cell, characterized by an abnormal proliferation of largely mature cells driven by mutations in JAK2, CALR, and MPL. All these mutations lead to a constitutive activation of the JAK-STAT signaling, which represents a target for therapy. Beyond driver ones, most patients, especially with myelofibrosis, harbor mutations in an array of "myeloid neoplasm-associated" genes that encode for proteins involved in chromatin modification and DNA methylation, RNA splicing, transcription regulation, and oncogenes. These additional mutations often arise in the context of clonal hematopoiesis of indeterminate potential (CHIP). The extensive characterization of the pathologic genome associated with MPN highlighted selected driver and non-driver mutations for their clinical informativeness. First, driver mutations are enlisted in the WHO classification as major diagnostic criteria and may be used for monitoring of residual disease after transplantation and response to treatment. Second, mutation profile can be used, eventually in combination with cytogenetic, histopathologic, hematologic, and clinical variables, to risk stratify patients regarding thrombosis, overall survival, and rate of transformation to secondary leukemia. This review outlines the molecular landscape of MPN and critically interprets current information for their potential impact on patient management.
Collapse
|
30
|
Nann D, Fend F. Synoptic Diagnostics of Myeloproliferative Neoplasms: Morphology and Molecular Genetics. Cancers (Basel) 2021; 13:cancers13143528. [PMID: 34298741 PMCID: PMC8303289 DOI: 10.3390/cancers13143528] [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: 06/09/2021] [Revised: 07/06/2021] [Accepted: 07/09/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary The diagnosis of myeloproliferative neoplasms requires assessment of a combination of clinical, morphological, immunophenotypic and genetic features, and this integrated, multimodal approach forms the basis for precise classification. Evaluation includes cell counts and morphology in the peripheral blood, bone marrow aspiration and trephine biopsy, and may encompass flow cytometry for specific questions. Diagnosis nowadays is completed by targeted molecular analysis for the detection of recurrent driver and, optionally, disease-modifying mutations. According to the current World Health Organization classification, all myeloproliferative disorders require assessment of molecular features to support the diagnosis or confirm a molecularly defined entity. This requires a structured molecular analysis workflow tailored for a rapid and cost-effective diagnosis. The review focuses on the morphological and molecular features of Ph-negative myeloproliferative neoplasms and their differential diagnoses, addresses open questions of classification, and emphasizes the enduring role of histopathological assessment in the molecular era. Abstract The diagnosis of a myeloid neoplasm relies on a combination of clinical, morphological, immunophenotypic and genetic features, and an integrated, multimodality approach is needed for precise classification. The basic diagnostics of myeloid neoplasms still rely on cell counts and morphology of peripheral blood and bone marrow aspirate, flow cytometry, cytogenetics and bone marrow trephine biopsy, but particularly in the setting of Ph− myeloproliferative neoplasms (MPN), the trephine biopsy has a crucial role. Nowadays, molecular studies are of great importance in confirming or refining a diagnosis and providing prognostic information. All myeloid neoplasms of chronic evolution included in this review, nowadays feature the presence or absence of specific genetic markers in their diagnostic criteria according to the current WHO classification, underlining the importance of molecular studies. Crucial differential diagnoses of Ph− MPN are the category of myeloid/lymphoid neoplasms with eosinophilia and gene rearrangement of PDGFRA, PDGFRB or FGFR1, or with PCM1-JAK2, and myelodysplastic/myeloproliferative neoplasms (MDS/MPN). This review focuses on morphological, immunophenotypical and molecular features of BCR-ABL1-negative MPN and their differential diagnoses. Furthermore, areas of difficulties and open questions in their classification are addressed, and the persistent role of morphology in the area of molecular medicine is discussed.
Collapse
Affiliation(s)
- Dominik Nann
- Institute of Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany;
- Comprehensive Cancer Center, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tübingen, 72076 Tübingen, Germany;
- Comprehensive Cancer Center, University Hospital Tübingen, 72076 Tübingen, Germany
- Correspondence: ; Tel.: +49-7071-2980207
| |
Collapse
|
31
|
Shide K. Calreticulin mutations in myeloproliferative neoplasms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 365:179-226. [PMID: 34756244 DOI: 10.1016/bs.ircmb.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Calreticulin (CALR) is a chaperone present in the endoplasmic reticulum, which is involved in the quality control of N-glycosylated proteins and storage of calcium ions. In 2013, the C-terminal mutation in CALR was identified in half of the patients with essential thrombocythemia and primary myelofibrosis who did not have a JAK2 or MPL mutation. The results of 8 years of intensive research are changing the clinical practice associated with treating myeloproliferative neoplasms (MPNs). The presence or absence of CALR mutations and their mutation types already provide important information for diagnosis and treatment decision making. In addition, the interaction with the thrombopoietin receptor MPL, which is the main mechanism of transformation by CALR mutation, and the expression of the mutant protein on the cell surface have a great potential as targets for molecular-targeted drugs and immunotherapy. This chapter presents recent findings on the clinical significance of the CALR mutation and the molecular basis by which this mutation drives MPNs.
Collapse
Affiliation(s)
- Kotaro Shide
- Division of Haematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
32
|
Induced Pluripotent Stem Cells Enable Disease Modeling and Drug Screening in Calreticulin del52 and ins5 Myeloproliferative Neoplasms. Hemasphere 2021; 5:e593. [PMID: 34131633 PMCID: PMC8196125 DOI: 10.1097/hs9.0000000000000593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Mutations in the calreticulin (CALR) gene are seen in about 30% of essential thrombocythemia and primary myelofibrosis patients. To address the contribution of the human CALR mutants to the pathogenesis of myeloproliferative neoplasms (MPNs) in an endogenous context, we modeled the CALRdel52 and CALRins5 mutants by induced pluripotent stem cell (iPSC) technology using CD34+ progenitors from 4 patients. We describe here the generation of several clones of iPSC carrying heterozygous CALRdel52 or CALRins5 mutations. We showed that CALRdel52 induces a stronger increase in progenitors than CALRins5 and that both CALRdel52 and CALRins5 mutants favor an expansion of the megakaryocytic lineage. Moreover, we found that both CALRdel52 and CALRins5 mutants rendered colony forming unit–megakaryocyte (CFU-MK) independent from thrombopoietin (TPO), and promoted a mild constitutive activation level of signal transducer and activator of transcription 3 in megakaryocytes. Unexpectedly, a mild increase in the sensitivity of colony forming unit-granulocyte (CFU-G) to granulocyte-colony stimulating factor was also observed in iPSC CALRdel52 and CALRins5 compared with control iPSC. Moreover, CALRdel52-induced megakaryocytic spontaneous growth is more dependent on Janus kinase 2/phosphoinositide 3-kinase/extracellular signal-regulated kinase than TPO-mediated growth and opens a therapeutic window for treatments in CALR-mutated MPN. The iPSC models described here represent an interesting platform for testing newly developed inhibitors. Altogether, this study shows that CALR-mutated iPSC recapitulate MPN phenotypes in vitro and may be used for drug screening.
Collapse
|
33
|
Activated IL-6 signaling contributes to the pathogenesis of, and is a novel therapeutic target for, CALR-mutated MPNs. Blood Adv 2021; 5:2184-2195. [PMID: 33890979 DOI: 10.1182/bloodadvances.2020003291] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 03/14/2021] [Indexed: 02/08/2023] Open
Abstract
Calreticulin (CALR), an endoplasmic reticulum-associated chaperone, is frequently mutated in myeloproliferative neoplasms (MPNs). Mutated CALR promotes downstream JAK2/STAT5 signaling through interaction with, and activation of, the thrombopoietin receptor (MPL). Here, we provide evidence of a novel mechanism contributing to CALR-mutated MPNs, represented by abnormal activation of the interleukin 6 (IL-6)-signaling pathway. We found that UT7 and UT7/mpl cells, engineered by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) to express the CALR type 1-like (DEL) mutation, acquired cytokine independence and were primed to the megakaryocyte (Mk) lineage. Levels of IL-6 messenger RNA (mRNA), extracellular-released IL-6, membrane-associated glycoprotein 130 (gp130), and IL-6 receptor (IL-6R), phosphorylated JAK1 and STAT3 (p-JAK1 and p-STAT3), and IL-6 promoter region occupancy by STAT3 all resulted in increased CALR DEL cells in the absence of MPL stimulation. Wild-type, but not mutated, CALR physically interacted with gp130 and IL-6R, downregulating their expression on the cell membrane. Agents targeting gp130 (SC-144), IL-6R (tocilizumab [TCZ]), and cell-released IL-6 reduced proliferation of CALR DEL as well as CALR knockout cells, supporting a mutated CALR loss-of-function model. CD34+ cells from CALR-mutated patients showed increased levels of IL-6 mRNA and p-STAT3, and colony-forming unit-Mk growth was inhibited by either SC144 or TCZ, as well as an IL-6 antibody, supporting cell-autonomous activation of the IL-6 pathway. Targeting IL-6 signaling also reduced colony formation by CD34+ cells of JAK2V617F-mutated patients. The combination of TCZ and ruxolitinib was synergistic at very low nanomolar concentrations. Overall, our results suggest that target inhibition of IL-6 signaling may have therapeutic potential in CALR, and possibly JAK2V617F, mutated MPNs.
Collapse
|
34
|
Acute promyelocyte leukemia arose from CALR 1 mutated post essential thrombocythemia- myelofibrosis with splanchnic vein thrombosis: A case report. Leuk Res Rep 2021; 15:100243. [PMID: 34040959 PMCID: PMC8142272 DOI: 10.1016/j.lrr.2021.100243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/19/2021] [Accepted: 05/02/2021] [Indexed: 11/23/2022] Open
Abstract
Major disease complications for patients with essential thrombocythemia (ET) include thrombosis and fibrotic or leukemic transformation. Calreticulin (CALR) mutation type 1 frequencies in ET are estimated between 7% and 11% and ET patients carrying CALR type 1 mutation are associated with lower risk of thrombosis but higher risk of myelofibrosis transformation compared to ET patients with JAK2 mutation. Leukemic transformation rates at 20 years are estimated at less than 5% for ET and risk factors for leukemic transformation are advanced age, thrombosis history, leukocytosis, and anemia. Amongst the subtypes of blast phase myeloproliferative neoplasms, acute promyelocytic leukemia is extremely rare. Herein, we present a case of a promyelocytic blast crisis of post-ET myelofibrosis with associated life-threatening splanchnic vein thrombosis. This case suggests that inflammation plays a key role in thrombotic events and fibrotic/leukemic transformation in ET patients, regardless the molecular landscape.
Collapse
|
35
|
Kanduła Z, Lewandowski K. Calreticulin – a multifaced protein. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.8892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Calreticulin (CALR) is a highly conserved multi-function protein that primarily localizes within
the lumen of the endoplasmic reticulum (ER). It participates in various processes in the cells,
including glycoprotein chaperoning, regulation of Ca2+ homeostasis, antigen processing and
presentation for adaptive immune response, cell adhesion/migration, cell proliferation, immunogenic
cell death, gene expression and RNA stability. The role of CALR in the assembly,
retrieval and cell surface expression of MHC class I molecules is well known. A fraction of
the total cellular CALR is localized in the cytosol, following its retro-translocation from the
ER. In the cell stress conditions, CALR is also expressed on the cell surface via an interaction
with phosphatidylserine localized on the inner leaflet of the plasma membrane. The abovementioned
mechanism is relevant for the recognition of the cells, as well as immunogenicity
and phagocytic uptake of proapoptotic and apoptotic cells.
Lastly, the presence of CALR exon 9 gene mutations was confirmed in patients with myeloproliferative
neoplasms. Their presence results in an abnormal CALR structure due to the
loss of its ER-retention sequence, CALR extra-ER localisation, the formation of a complex
with thrombopoietin receptor, and oncogenic transformation of hematopoietic stem cells. It
is also known that CALR exon 9 mutants are highly immunogenic and induce T cell response.
Despite this fact, CALR mutant positive hematopoietic cells emerge. The last phenomenon is
probably the result of the inhibition of phagocytosis of the cancer cells exposing CALR mutant
protein by dendritic cells.
Collapse
Affiliation(s)
- Zuzanna Kanduła
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poland
| | - Krzysztof Lewandowski
- Department of Hematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poland
| |
Collapse
|
36
|
Mutations and thrombosis in essential thrombocythemia. Blood Cancer J 2021; 11:77. [PMID: 33907189 PMCID: PMC8079671 DOI: 10.1038/s41408-021-00470-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 03/04/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
|
37
|
Benlabiod C, Dagher T, Marty C, Villeval JL. Lessons from mouse models of MPN. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 366:125-185. [PMID: 35153003 DOI: 10.1016/bs.ircmb.2021.02.009] [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] [Indexed: 06/14/2023]
Abstract
Over the past decades, a variety of MPN mouse models have been developed to express in HSC the main mutations identified in patients: JAK2V617F, CALRdel52 or ins5 and MPLW515L. These models mimic quite faithfully human PV or ET with their natural evolutions into MF and their hemostasis complications, demonstrating the driver function of these mutations in MPN. Here, we review these models and show how they have improved our general understanding of MPN regarding (1) the mechanisms of fibrosis, thrombosis/hemorrhages and disease initiation, (2) the roles of additional mutations and signaling pathways in disease progression and (3) the preclinical development of novel therapies. We also address controversial results between these models and remind how these models may differ from human MPN onset and also how basically mice are not humans, encouraging caution when one draw lessons from mice to humans. Furthermore, the contribution of germline genetic predisposition, HSC and niche aging, metabolic, oxidative, replicative or genotoxic stress, inflammation, immune escape and additional mutations need to be considered in further investigations to encompass the full complexity of human MPN in mice.
Collapse
Affiliation(s)
- Camelia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France
| | - Tracy Dagher
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France.
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France; Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France; Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
38
|
Gangat N, Szuber N, Jawaid T, Hanson CA, Pardanani A, Tefferi A. Young platelet millionaires with essential thrombocythemia. Am J Hematol 2021; 96:E93-E95. [PMID: 33524176 DOI: 10.1002/ajh.26114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Naseema Gangat
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota
| | - Natasha Szuber
- Department of Hematology Université de Montréal Montréal Quebec Canada
| | - Tabinda Jawaid
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota
| | - Curtis A. Hanson
- Division of Hematopathology, Department of Laboratory Medicine Mayo Clinic Rochester Minnesota
| | - Animesh Pardanani
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota
| | - Ayalew Tefferi
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota
| |
Collapse
|
39
|
Mutation profile in BCR-ABL1-negative myeloproliferative neoplasms: A single-center experience from India. Hematol Oncol Stem Cell Ther 2021; 15:13-20. [PMID: 33789164 DOI: 10.1016/j.hemonc.2021.03.002] [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: 01/07/2021] [Accepted: 03/06/2021] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE/BACKGROUND Recurrent somatic mutations in the JAK2, calreticulin (CALR), and the MPL genes are described as drivers of BCR-ABL1-negative myeloproliferative neoplasms (MPN) that includes polycythemia vera (PV), essential thrombocytosis (ET), primary myelofibrosis (PMF), and MPN unclassified (MPN-U). METHODS We describe the mutation profile and clinical features of MPN cases diagnosed at a tertiary care center. JAK2V617F and MPL (S505/W515) mutations were screened by allele-specific polymerase chain reaction, while CALR exon 9 and JAK2 exon 12 mutations were screened by fragment analysis/Sanger sequencing. Among the 1,570 patients tested for these mutations during the study period, 407 were classified as MPN with a diagnosis of PV, ET, PMF, and MPN-U seen in 30%, 17%, 36%, and 17%, respectively, screened. RESULTS Similar to previous reports from Asian countries, the incidence of PMF was the highest among the classic MPN. JAK2V617F mutation was detected in 90% of PV, 38% of ET, 48% of PMF, and 65% of MPN-U. JAK2 exon 12 mutations were seen in 5.7% of PV and 1.4% of PMF. CALR exon 9 mutations were seen in 33% of ET, 33% of PMF, and 12% of MPN-U. MPL mutations were detected in 2.8%, 2.7%, and 2.9% of ET, PMF, and MPN-U, respectively. Fifteen % of PMF, 26% of ET, and 22% of MPN-U were triple negative. CONCLUSION There was a significantly higher incidence of CALR mutation in PMF and ET cases. Our study highlights the challenges in the diagnosis of JAK2-negative PV and the need for harmonization of criteria for the same.
Collapse
|
40
|
Cattaneo D, Croci GA, Bucelli C, Tabano S, Cannone MG, Gaudioso G, Barbanti MC, Barbullushi K, Bianchi P, Fermo E, Fabris S, Baldini L, Gianelli U, Iurlo A. Triple-Negative Essential Thrombocythemia: Clinical-Pathological and Molecular Features. A Single-Center Cohort Study. Front Oncol 2021; 11:637116. [PMID: 33791220 PMCID: PMC8006378 DOI: 10.3389/fonc.2021.637116] [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: 12/02/2020] [Accepted: 02/18/2021] [Indexed: 02/02/2023] Open
Abstract
Lack of demonstrable mutations affecting JAK2, CALR, or MPL driver genes within the spectrum of BCR-ABL1-negative myeloproliferative neoplasms (MPNs) is currently referred to as a triple-negative genotype, which is found in about 10% of patients with essential thrombocythemia (ET) and 5-10% of those with primary myelofibrosis (PMF). Very few papers are presently available on triple-negative ET, which is basically described as an indolent disease, differently from triple-negative PMF, which is an aggressive myeloid neoplasm, with a significantly higher risk of leukemic evolution. The aim of the present study was to evaluate the bone marrow morphology and the clinical-laboratory parameters of triple-negative ET patients, as well as to determine their molecular profile using next-generation sequencing (NGS) to identify any potential clonal biomarkers. We evaluated a single-center series of 40 triple-negative ET patients, diagnosed according to the 2017 WHO classification criteria and regularly followed up at the Hematology Unit of our Institution, between January 1983 and January 2019. In all patients, NGS was performed using the Illumina Ampliseq Myeloid Panel; morphological and immunohistochemical features of the bone marrow trephine biopsies were also thoroughly reviewed. Nucleotide variants were detected in 35 out of 40 patients. In detail, 29 subjects harbored one or two variants and six cases showed three or more concomitant nucleotide changes. The most frequent sequence variants involved the TET2 gene (55.0%), followed by KIT (27.5%). Histologically, most of the cases displayed a classical ET morphology. Interestingly, prevalent megakaryocytes morphology was more frequently polymorphic with a mixture of giant megakaryocytes with hyperlobulated nuclei, normal and small sized maturing elements, and naked nuclei. Finally, in five cases a mild degree of reticulin fibrosis (MF-1) was evident together with an increase in the micro-vessel density. By means of NGS we were able to identify nucleotide variants in most cases, thus we suggest that a sizeable proportion of triple-negative ET patients do have a clonal disease. In analogy with driver genes-mutated MPNs, these observations may prevent issues arising concerning triple-negative ET treatment, especially when a cytoreductive therapy may be warranted.
Collapse
Affiliation(s)
- Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giorgio Alberto Croci
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Division of Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Tabano
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Laboratory of Medical Genetics, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Giulia Cannone
- Laboratory of Medical Genetics, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Dermatology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriella Gaudioso
- Division of Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Chiara Barbanti
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Kordelia Barbullushi
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Paola Bianchi
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elisa Fermo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sonia Fabris
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Baldini
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Umberto Gianelli
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Division of Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
41
|
Ross DM, Thomson C, Hamad N, Lane SW, Manos K, Grigg AP, Guo B, Erber WN, Scott A, Viiala N, Chee L, Latimer M, Tate C, Grove C, Perkins AC, Blombery P. Myeloid somatic mutation panel testing in myeloproliferative neoplasms. Pathology 2021; 53:339-348. [PMID: 33674147 DOI: 10.1016/j.pathol.2021.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/14/2021] [Accepted: 01/17/2021] [Indexed: 12/22/2022]
Abstract
Myeloproliferative neoplasms are characterised by somatic mutations in pathways that regulate cell proliferation, epigenetic modifications, RNA splicing or DNA repair. Assessment of the mutational profile assists diagnosis and classification, but also aids assessment of prognosis, and may guide the use of emerging targeted therapies. The most practical way to provide information on numerous genetic variants is by using massively parallel sequencing, commonly in the form of disease specific next generation sequencing (NGS) panels. This review summarises the diagnostic and prognostic value of somatic mutation testing in Philadelphia-negative myeloproliferative neoplasms: polycythaemia vera, essential thrombocythaemia, primary myelofibrosis, chronic neutrophilic leukaemia, systemic mastocytosis, and chronic eosinophilic leukaemia. NGS panel testing is increasing in routine practice and promises to improve the accuracy and efficiency of pathological diagnosis and prognosis.
Collapse
Affiliation(s)
- David M Ross
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, University of Adelaide, Adelaide, SA, Australia; Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia; Department of Haematology and Genetic Pathology, Flinders University and Medical Centre, Adelaide, SA, Australia.
| | - Candice Thomson
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, University of Adelaide, Adelaide, SA, Australia
| | - Nada Hamad
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Haematology Department, St Vincent's Hospital, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Steven W Lane
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia; QIMR Berghofer Medical Research Institute, University of Queensland, Brisbane, Qld, Australia
| | - Kate Manos
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Clinical Haematology, Austin Health, Heidelberg, Vic, Australia
| | - Andrew P Grigg
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Clinical Haematology, Austin Health, Heidelberg, Vic, Australia
| | - Belinda Guo
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Wendy N Erber
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia; Haematology Department, PathWest Laboratory Medicine, Perth, WA, Australia
| | - Ashleigh Scott
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Haematology and Bone Marrow Transplantation, Royal Brisbane and Women's Hospital, Brisbane, Qld, Australia
| | - Nick Viiala
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Haematology, Liverpool Hospital, University of New South Wales, Sydney, NSW, Australia
| | - Lynette Chee
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Clinical Haematology, Royal Melbourne Hospital, Peter MacCallum Cancer Centre, Department of Medicine, The University of Melbourne, Melbourne, Vic, Australia
| | - Maya Latimer
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; ACT Pathology and Canberra Hospital, Australian National University, Canberra, ACT, Australia
| | - Courtney Tate
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Haematology Department, Gold Coast University Hospital, University of Queensland, Southport, Qld, Australia
| | - Carolyn Grove
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia; Haematology Department, PathWest Laboratory Medicine, Perth, WA, Australia; Haematology Department, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Andrew C Perkins
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Haematology, Alfred Hospital, Monash University, Melbourne, Vic, Australia
| | - Piers Blombery
- Myeloproliferative Neoplasms Working Party, Australasian Leukaemia and Lymphoma Group, Melbourne, Vic, Australia; Department of Clinical Haematology, Royal Melbourne Hospital, Peter MacCallum Cancer Centre, Department of Medicine, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
42
|
Yamade K, Yamaguchi T, Nagai Y, Kamisako T. Platelet count evaluation compared with the immunoplatelet reference method and performance evaluation of the hematology analyzer Celltac G. Int J Lab Hematol 2021; 43:927-938. [PMID: 33548102 DOI: 10.1111/ijlh.13481] [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: 08/25/2020] [Revised: 12/14/2020] [Accepted: 01/11/2021] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The hematology analyzer, Celltac G (Nihon Kohden), designed to improve platelet count (Plt) accuracy, is equipped with new sheath flow control technology. Clinical evaluation of the Celltac G was assessed by comparability with XN-9000 (Sysmex Corporation) and CELL-DYN Sapphire (Abbott Diagnostics). The accuracy of all three analyzers, which use different measuring principles, was compared with the immunoplatelet reference method (FCM-Ref). METHODS Repeatability and within-laboratory imprecision were assessed using 10 clinical fresh whole blood samples and three control materials with differing levels. Carryover was evaluated using 6 clinical fresh whole blood samples. For method comparison between the three analyzers, 388 samples were used. Plt accuracy among the three analyzers was evaluated using 54 blood samples, including 42 samples with a platelet count less than 50x109 /L. The International Council for Standardization in Haematology method for Plt was used as the FCM-Ref. RESULTS The Celltac G showed sufficient performance with regard to imprecision, carryover, and comparability. The Analytical Measurement Interval (AMI) and linearity for all parameters of Plt were validated within 4.6 to 809.1 (×109 /L). All hematology analyzers showed some disagreement in Plt when compared with the immunoplatelet reference method. CONCLUSION The Celltac G hematology analyzer is suitable for clinical use. Platelet count evaluation of the three analyzers suggests the need to determine a reportable measurement interval (RMI) in the clinical laboratory for adequate reporting of a Plt from multiple different values.
Collapse
Affiliation(s)
- Kenji Yamade
- Department of Central Clinical Laboratory, Kindai University Hospital, Osakasayama, Japan.,Kindai University Graduate School of Medical Sciences, Osakasayama, Japan
| | - Toshihiro Yamaguchi
- Department of Central Clinical Laboratory, Kindai University Hospital, Osakasayama, Japan
| | - Yutaka Nagai
- Faculty of Clinical Laboratory, Kansai University of Health Sciences, Kumatori, Japan.,IVD Business Operations, Nihon Kohden Corp., Tokyo, Japan
| | - Toshinori Kamisako
- Department of Central Clinical Laboratory, Kindai University Hospital, Osakasayama, Japan.,Kindai University Graduate School of Medical Sciences, Osakasayama, Japan
| |
Collapse
|
43
|
Safavi M, Monabati A, Safaei A, Mirtalebi MS, Faghih M. JAK2, CALR, and MPL Mutation Profiles in BCR-ABL Negative Myeloproliferative Neoplasms, a Referral Center Experience in the Middle East. IRANIAN JOURNAL OF PATHOLOGY 2021; 16:190-194. [PMID: 33936230 PMCID: PMC8085288 DOI: 10.30699/ijp.2021.136458.2495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/10/2021] [Indexed: 11/25/2022]
Abstract
Background & Objective: JAK2, CALR, and MPL genes play pivotal roles in the pathogenesis of BCR-ABL negative myeloproliferative neoplasms. This study was conducted to evaluate the frequency of JAK2, CALR, and MPL mutations in BCR-ABL negative myeloproliferative neoplasms and their association with demographic data and hematologic parameters in a referral center, in the Middle East. Methods: Seventy-one patients with BCR-ABL negative myeloproliferative neoplasms were evaluated for JAK2 V617F, CALR type 1, CALR type 2, and MPL by allele-specific PCR and conventional PCR from 2018 to 2019. Results: Twenty-three patients were categorized as polycythemia vera, JAK2 V617F was observed in 91.3% of these cases. Thirty-eight patients were classified as essential thrombocythemia of which 52.6% showed JAK2 V617F, 18.4% demonstrated CALR type 1, 7.9% denoted CALR type 2 and there was no mutation reported in 21.1%. Seven patients were recognized as primary myelofibrosis and exhibited JAK2 V617F mutation in 57.1%, CALR type 1 in 14.3 %, CALR type 2 in 14.3% and no mutation in 14.3%. Three patients were diagnosed as MPN, unclassifiable and 33.3% revealed JAK2 V617F mutation, and no mutation was found in 66.6%. The age (59.15±13.10) and neutrophil percent (70.78±10.14) were higher in patients with JAK2 V617 mutation compared to other mutations (P=0.000, and P=0.03). Platelet count was significantly higher in patients with CALR type 1 mutation (1240400± 402053) (P=0.000). Conclusion: JAK2 V617F was associated with patients’ higher age and higher neutrophil count in CBC. CALR mutation had an association with higher platelet count. No MPL mutation was found in this study and it seems that its frequency is lower than what is expected in this region.
Collapse
Affiliation(s)
- Moeinadin Safavi
- Molecular Pathology and Cytogenetic Section, Department of Pathology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Monabati
- Molecular Pathology and Cytogenetic Section, Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Akbar Safaei
- Molecular Pathology and Cytogenetic Section, Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Sadat Mirtalebi
- Molecular Pathology and Cytogenetic Section, Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoumeh Faghih
- Molecular Pathology and Cytogenetic Section, Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
44
|
Morishita S, Ochiai T, Misawa K, Osaga S, Inano T, Fukuda Y, Edahiro Y, Ohsaka A, Araki M, Komatsu N. Clinical impacts of the mutational spectrum in Japanese patients with primary myelofibrosis. Int J Hematol 2021; 113:500-507. [PMID: 33389584 DOI: 10.1007/s12185-020-03054-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/26/2020] [Accepted: 11/26/2020] [Indexed: 11/26/2022]
Abstract
Patients with primary myelofibrosis (PMF) have a poorer prognosis than those with other subtypes of myeloproliferative neoplasms (MPNs). To investigate the relationship between gene mutations and the prognosis of Japanese PMF patients, we analyzed mutations in 72 regions located in 14 MPN-relevant genes (CSF3R, MPL, JAK2, CALR, DNMT3A, TET2, EZH2, ASXL1, IDH1/2, SRSF2, SF3B1, U2AF1, and TP53) utilizing a target resequencing platform. In our cohort, ASXL1 mutations were more frequently detected in both overt and prefibrotic PMF patients than other mutations. The frequency of ASXL1 mutations was slightly higher among overt PMF patients than among prefibrotic PMF patients (44.6% vs 25.0%, FDR = 0.472). Decision tree classification algorithms revealed that ASXL1, EZH2, and SRSF2 mutations were associated with a poor prognosis for overt PMF. Overall survival was significantly shorter in patients harboring ASXL1, EZH2, or SRSF2 mutations than in those without these mutations (p = 0.03). These results suggest that, as reported in Western countries, MIPSS70 is applicable to Japanese PMF patients and ASXL1, EZH2, and SRSF2 mutations may be utilized as surrogate markers of a poor prognosis.
Collapse
Affiliation(s)
- Soji Morishita
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Tomonori Ochiai
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kyohei Misawa
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Satoshi Osaga
- Clinical Research Management Center, Nagoya City University Hospital, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Tadaaki Inano
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yasutaka Fukuda
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yoko Edahiro
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Akimichi Ohsaka
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Marito Araki
- Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
45
|
Pérez Encinas MM, Sobas M, Gómez-Casares MT, Abuin Blanco A, Noya Pereira MS, Raya JM, Andrade-Campos MM, Álvarez Larrán A, Lewandowski K, Łukasz S, Hernández Boluda JC, Ferrer-Marín F, Fox ML, Gołos A, Gasior Kabat M, Magro Mazo E, Czyż A, Martín Martín A, Bellosillo Paricio B, Quinteiro García C, González Martín JM, Stuckey R. The risk of thrombosis in essential thrombocythemia is associated with the type of CALR mutation: A multicentre collaborative study. Eur J Haematol 2020; 106:371-379. [PMID: 33275803 DOI: 10.1111/ejh.13561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES In patients with essential thrombocythemia (ET), after the JAK2V617F driver mutation, mutations in CALR are common (classified as type 1, 52-bp deletion or type 2, 5-bp insertion). CALR mutations have generally been associated with a lower risk of thrombosis. This study aimed to confirm the impact of CALR mutation type on thrombotic risk. METHODS We retrospectively investigated 983 ET patients diagnosed in Spanish and Polish hospitals. RESULTS With 7.5 years of median follow-up from diagnosis, 155 patients (15.8%) had one or more thrombotic event. The 5-year thrombosis-free survival (TFS) rate was 83.8%, 91.6% and 93.9% for the JAK2V617F, CALR-type 1 and CALR-type 2 groups, respectively (P = .002). Comparing CALR-type 1 and CALR-type 2 groups, TFS for venous thrombosis was lower in CALR-type 1 (P = .046), with no difference in TFS for arterial thrombosis observed. The cumulative incidence of thrombosis was significantly different comparing JAK2V617F vs CALR-type 2 groups but not JAK2V617F vs CALR-type 1 groups. Moreover, CALR-type 2 mutation was a statistically significant protective factor for thrombosis with respect to JAK2V617F in multivariate logistic regression (OR: 0.45, P = .04) adjusted by age. CONCLUSIONS Our results suggest that CALR mutation type has prognostic value for the stratification of thrombotic risk in ET patients.
Collapse
Affiliation(s)
- Manuel M Pérez Encinas
- Hematology Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Marta Sobas
- Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - María Teresa Gómez-Casares
- Hematology Department, Hospital Universitario de Gran Canaria Dr.Negrín, Las Palmas de Gran Canaria, Spain
| | - Aitor Abuin Blanco
- Hematology Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | | | - José María Raya
- Hematology Department, Hospital, Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | | | | | - Krzysztof Lewandowski
- Department of Hematology and Bone Marrow Transplantation, University of Medical Sciences, Poznan, Poland
| | - Szukalski Łukasz
- Department of Hematology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Torun, Poland
| | | | | | - María Laura Fox
- Hematology Department, Hospital Vall d'Hebron, Barcelona, Spain
| | - Aleksandra Gołos
- Department of Clinical Oncology and Chemotherapy, Magodent Hospital, Warsaw, Poland
| | | | - Elena Magro Mazo
- Hematology Department, Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Anna Czyż
- Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | | | | | - Celsa Quinteiro García
- Genomics Laboratory, Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela, Spain
| | | | - Ruth Stuckey
- Hematology Department, Hospital Universitario de Gran Canaria Dr.Negrín, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
46
|
Tefferi A, Barbui T. Polycythemia vera and essential thrombocythemia: 2021 update on diagnosis, risk-stratification and management. Am J Hematol 2020; 95:1599-1613. [PMID: 32974939 DOI: 10.1002/ajh.26008] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/25/2022]
Abstract
DISEASE OVERVIEW Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPN) respectively characterized by clonal erythrocytosis and thrombocytosis; other disease features include leukocytosis, splenomegaly, thrombosis, bleeding, microcirculatory symptoms, pruritus and risk of leukemic or fibrotic transformation. DIAGNOSIS Bone marrow morphology remains the cornerstone of diagnosis. In addition, the presence of JAK2 mutation is expected in PV while approximately 90% of patients with ET express mutually exclusive JAK2, CALR or MPL mutations (so called driver mutations). In ET, it is most important to exclude the possibility of prefibrotic myelofibrosis. SURVIVAL Median survivals are approximately 15 years for PV and 18 years for ET; the corresponding values for patients age 40 or younger were 37 and 35 years. Certain mutations (mostly spliceosome) and abnormal karyotype might compromise survival in PV and ET. Life-expectancy in ET is inferior to the control population. Driver mutations have not been shown to affect survival in ET but risk of thrombosis is higher in JAK2 mutated cases. Leukemic transformation rates at 10 years are estimated at <1% for ET and 3% for PV. THROMBOSIS RISK In PV, two risk categories are considered: high (age > 60 years or thrombosis history present) and low (absence of both risk factors). In ET, four risk categories are considered: very low (age ≤ 60 years, no thrombosis history, JAK2 wild-type), low (same as very low but JAK2 mutation present), intermediate (age > 60 years, no thrombosis history, JAK2 wild-type) and high (thrombosis history present or age > 60 years with JAK2 mutation). RISK-ADAPTED THERAPY The main goal of therapy in both PV and ET is to prevent thrombohemorrhagic complications. All patients with PV require phlebotomy to keep hematocrit below 45% and once-daily or twice-daily aspirin (81 mg), in the absence of contraindications. Very low risk ET might not require therapy while aspirin therapy is advised for low risk disease. Cytoreductive therapy is recommended for high-risk ET and PV, but it is not mandatory for intermediate-risk ET. First-line drug of choice for cytoreductive therapy, in both ET and PV, is hydroxyurea and second-line drugs of choice are interferon-α and busulfan. We do not recommend treatment with ruxolutinib in PV, unless in the presence of severe and protracted pruritus or marked splenomegaly that is not responding to the aforementioned drugs. NEW TREATMENT DIRECTIONS Controlled studies are needed to confirm the clinical outcome value of twice-daily vs once-daily aspirin dosing and the therapeutic role of pegylated interferons and direct oral anticoagulants.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota
| | - Tiziano Barbui
- Research Foundation Papa Giovanni XXIII Hospital Bergamo Italy
| |
Collapse
|
47
|
Loscocco GG, Guglielmelli P, Vannucchi AM. Impact of Mutational Profile on the Management of Myeloproliferative Neoplasms: A Short Review of the Emerging Data. Onco Targets Ther 2020; 13:12367-12382. [PMID: 33293830 PMCID: PMC7718985 DOI: 10.2147/ott.s287944] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/18/2020] [Indexed: 12/16/2022] Open
Abstract
Philadelphia-chromosome negative myeloproliferative neoplasms (MPN) are a heterogeneous group of clonal hematopoietic stem cell disorders characterized by an increased risk of thrombosis and progression to acute myeloid leukemia. MPN are associated with driver mutations in JAK2, CALR and MPL which are crucial for the diagnosis and lead to a constitutive activation of the JAK-STAT signaling, independent of cytokine regulation. Moreover, most patients have concomitant mutations in genes involved in DNA methylation, chromatin modification, messenger RNA splicing, transcription regulation and signal transduction. These additional mutations may arise before, in the context of clonal hematopoiesis of indeterminate potential (CHIP), or after the acquisition of the driver mutation. The clinical phenotype of MPN results from complex interactions between mutations and host factors. The increased application of next-generation sequencing (NGS) techniques to a large series of patients with MPN has expanded the knowledge of mutational landscape and contributed to define the clinical significance of mutations. This molecular information is being increasingly used to refine diagnosis, risk stratification, monitoring of residual disease and response to treatment. ASXL1, SRSF2, EZH2, IDH1/IDH2 and U2AF1 mutations are associated with a more advanced disease and reduced overall survival in primary myelofibrosis (PMF), whereas spliceosome mutations in Polycythemia vera (PV) and essential thrombocythemia (ET) adversely affect both overall (SF3B1, SRSF2 in ET and SRSF2 in PV) and myelofibrosis-free (U2AF1, SF3B1 in ET) survival. This review discusses current knowledge of the molecular landscape of MPN, and how the availability of those molecular information may impact patient management.
Collapse
Affiliation(s)
- Giuseppe G Loscocco
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliero-Universitaria Careggi, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paola Guglielmelli
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliero-Universitaria Careggi, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandro M Vannucchi
- CRIMM, Centro di Ricerca e Innovazione per le Malattie Mieloproliferative, Azienda Ospedaliero-Universitaria Careggi, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
48
|
Benlabiod C, Cacemiro MDC, Nédélec A, Edmond V, Muller D, Rameau P, Touchard L, Gonin P, Constantinescu SN, Raslova H, Villeval JL, Vainchenker W, Plo I, Marty C. Calreticulin del52 and ins5 knock-in mice recapitulate different myeloproliferative phenotypes observed in patients with MPN. Nat Commun 2020; 11:4886. [PMID: 32985500 PMCID: PMC7522233 DOI: 10.1038/s41467-020-18691-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Somatic mutations in the calreticulin (CALR) gene are associated with approximately 30% of essential thrombocythemia (ET) and primary myelofibrosis (PMF). CALR mutations, including the two most frequent 52 bp deletion (del52) and 5 bp insertion (ins5), induce a frameshift to the same alternative reading frame generating new C-terminal tails. In patients, del52 and ins5 induce two phenotypically distinct myeloproliferative neoplasms (MPNs). They are equally found in ET, but del52 is more frequent in PMF. We generated heterozygous and homozygous conditional inducible knock-in (KI) mice expressing a chimeric murine CALR del52 or ins5 with the human mutated C-terminal tail to investigate their pathogenic effects on hematopoiesis. Del52 induces greater phenotypic changes than ins5 including thrombocytosis, leukocytosis, splenomegaly, bone marrow hypocellularity, megakaryocytic lineage amplification, expansion and competitive advantage of the hematopoietic stem cell compartment. Homozygosity amplifies these features, suggesting a distinct contribution of homozygous clones to human MPNs. Moreover, homozygous del52 KI mice display features of a penetrant myelofibrosis-like disorder with extramedullary hematopoiesis linked to splenomegaly, megakaryocyte hyperplasia and the presence of reticulin fibers. Overall, modeling del52 and ins5 mutations in mice successfully recapitulates the differences in phenotypes observed in patients. Calreticulin del52 and ins5 mutations induce two phenotypically distinct myeloproliferative neoplasms in patients. Here the authors show that modeling these mutations in knock-in mice recapitulate the two diseases and highlight how they impact the different hematopoietic compartments.
Collapse
Affiliation(s)
- Camélia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Maira da Costa Cacemiro
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France.,Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Audrey Nédélec
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Valérie Edmond
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Delphine Muller
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Philippe Rameau
- Integrated Biology Core Facility, Gustave Roussy, Villejuif, France
| | - Laure Touchard
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Patrick Gonin
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France. .,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France. .,Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
49
|
Jacquelin S, Kramer F, Mullally A, Lane SW. Murine Models of Myelofibrosis. Cancers (Basel) 2020; 12:cancers12092381. [PMID: 32842500 PMCID: PMC7563264 DOI: 10.3390/cancers12092381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 01/22/2023] Open
Abstract
Myelofibrosis (MF) is subtype of myeloproliferative neoplasm (MPN) characterized by a relatively poor prognosis in patients. Understanding the factors that drive MF pathogenesis is crucial to identifying novel therapeutic approaches with the potential to improve patient care. Driver mutations in three main genes (janus kinase 2 (JAK2), calreticulin (CALR), and myeloproliferative leukemia virus oncogene (MPL)) are recurrently mutated in MPN and are sufficient to engender MPN using animal models. Interestingly, animal studies have shown that the underlying molecular mutation and the acquisition of additional genetic lesions is associated with MF outcome and transition from early stage MPN such as essential thrombocythemia (ET) and polycythemia vera (PV) to secondary MF. In this issue, we review murine models that have contributed to a better characterization of MF pathobiology and identification of new therapeutic opportunities in MPN.
Collapse
Affiliation(s)
- Sebastien Jacquelin
- Cancer program QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Correspondence: (S.J.); (S.W.L.)
| | - Frederike Kramer
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.K.); (A.M.)
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (F.K.); (A.M.)
| | - Steven W. Lane
- Cancer program QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4006, Australia
- Cancer Care Services, The Royal Brisbane and Women’s Hospital, Brisbane 4029, Australia
- University of Queensland, St Lucia, QLD 4072, Australia
- Correspondence: (S.J.); (S.W.L.)
| |
Collapse
|
50
|
Different impact of calreticulin mutations on human hematopoiesis in myeloproliferative neoplasms. Oncogene 2020; 39:5323-5337. [PMID: 32572159 DOI: 10.1038/s41388-020-1368-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/05/2020] [Accepted: 06/11/2020] [Indexed: 01/05/2023]
Abstract
Mutations of calreticulin (CALRm) define a subtype of myeloproliferative neoplasms (MPN). We studied the biological and genetic features of CALR-mutated essential thrombocythemia and myelofibrosis patients. In most cases, CALRm were found in granulocytes, monocytes, B and NK cells, but also in T cells. However, the type 1 CALRm spreads more easily than the type 2 CALRm in lymphoid cells. The CALRm were also associated with an early clonal dominance at the level of hematopoietic stem and progenitor cells (HSPC) with no significant increase during granulo/monocytic differentiation in most cases. Moreover, we found that half of type 2 CALRm patients harbors some homozygous progenitors. Those patients were associated with a higher clonal dominance during granulo/monocytic differentiation than patients with only heterozygous type 2 CALRm progenitors. When associated mutations were present, CALRm were the first genetic event suggesting that they are both the initiating and phenotypic event. In blood, type 1 CALRm led to a greater increased number of all types of progenitors compared with the type 2 CALRm. However, both types of CALRm induced an increase in megakaryocytic progenitors associated with a ruxolitinib-sensitive independent growth and with a mild constitutive signaling in megakaryocytes. At the transcriptional level, type 1 CALRm seems to deregulate more pathways than the type 2 CALRm in megakaryocytes. Altogether, our results show that CALRm modify both the HSPC and megakaryocyte biology with a stronger effect for type 1 than for type 2 CALRm.
Collapse
|