1
|
Reiter A, Metzgeroth G, Cross NCP. How I diagnose and treat myeloid/lymphoid neoplasms with tyrosine kinase gene fusions. Blood 2025; 145:1758-1768. [PMID: 39046810 DOI: 10.1182/blood.2023022417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/03/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024] Open
Abstract
ABSTRACT The fifth edition of the World Health Organization (WHO) classification and the International Consensus Classification (ICC) both include a category "myeloid/lymphoid neoplasms (MLN) with eosinophilia (eo) and tyrosine kinase (TK) gene fusions" (WHO, MLN-TK; ICC, M/LN-eo-TK). This rare group comprises phenotypically and prognostically heterogeneous disorders, which present a significant diagnostic challenge. The rapid and reliable identification of patients with MLN-TK may be delayed due to genetic complexity and significant phenotypic differences, including the chronic phase and primary/secondary blast phase (BP) of myeloid, lymphoid, or mixed phenotype in the bone marrow (BP-BM) and/or at extramedullary sites (extramedullary disease [EMD]). As a result, the entire armamentarium of conventional molecular genetic and cytogenetic techniques complemented by modern sequencing technologies, such as RNA sequencing or whole-genome sequencing, are often required to identify an underlying TK fusion. TK inhibitors (TKIs) with variable efficacy are available for all fusion genes, but a long-term favorable clinical course under TKI monotherapy is currently only observed in MLN-PDGFRA/PDGFRB fusion genes on imatinib. Because primary/secondary BP-BM/EMD occurs more frequently in MLN-FGFR1/JAK2/FLT3/ETV6::ABL1, a sequential combination of selective TKIs with or without prior intensive chemotherapy, rarely local radiotherapy, and/or subsequent allogeneic hematopoietic cell transplantation should be considered.
Collapse
Affiliation(s)
- Andreas Reiter
- Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Georgia Metzgeroth
- Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Nicholas C P Cross
- Wessex Genomics Laboratory Service, Salisbury, United Kingdom
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
2
|
Mlayah Z, Ben-Rekaya I, Bizid I, Slama N, Boukhris S, Laatiri MA. Clonal eosinophilia with exclusive pulmonary involvement driven by PDGFRA rearrangement treated with imatinib: A case report. Leuk Res Rep 2025; 23:100502. [PMID: 40115037 PMCID: PMC11925588 DOI: 10.1016/j.lrr.2025.100502] [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: 12/12/2024] [Revised: 02/02/2025] [Accepted: 02/12/2025] [Indexed: 03/22/2025] Open
Abstract
Hypereosinophilic syndrome (HES) was first described in 1968 by Hardy and Anderson. It is a group of rare, multisystemic and heterogeneous pathologies, characterized by significant morbidity and mortality. The occurrence of clonal hypereosinophilic syndrome associated with FIP 1L1-PDGFRA+ is estimated to range between 0.31 and 6.3 cases per million individuals. The organs most commonly impacted are the heart and spleen, with the lungs being the next most affected. Clonal hypereosinophilic syndromes with exclusive pulmonary involvement are exceptional. Due to the rarity of clonal HES, this report aims to not only describe the patient's clinical, biological, and radiological manifestations of clonal HES but also enrich the literature to ameliorate the management of this uncommon syndrome. we report the case of a patient with past medical history of obstructive bronchopneumopathy who was presented with cough and dyspnea. Investigations revealed peripheral blood hypereosinophilia (between 4000 and 9000/mm3) which lead us to suspect clonal hypereosinophilic syndrome (HES). This diagnosis was confirmed by cytogenetics/fluorescence in situ hybridization (FISH) which demonstrated a positive FIP 1L1-PDGFRA rearrangement. The CTAP confirmed isolated lung involvement with interstitial infiltrate of the subpleural territories of both lung bases and the bronchoalveolar lavage showed eosinophil count elevated at 15%. The patient was treated by imatinib at a dose of 100 mg/day was initiated. The patient follow-up showed a reduction in eosinophils count to 7500/mm3 at two months of treatment. A molecular evaluation is scheduled in 3 months to assess the response to imatinib.
Collapse
Affiliation(s)
- Zaineb Mlayah
- Clinical hematology department of Fattouma Bourguiba Hospital, Monatir, Tunisia
| | - Inés Ben-Rekaya
- Clinical hematology department of Fattouma Bourguiba Hospital, Monatir, Tunisia
| | - Inaam Bizid
- Clinical hematology department of Fattouma Bourguiba Hospital, Monatir, Tunisia
| | - Nader Slama
- Clinical hematology department of Fattouma Bourguiba Hospital, Monatir, Tunisia
| | - Sara Boukhris
- Clinical hematology department of Fattouma Bourguiba Hospital, Monatir, Tunisia
| | | |
Collapse
|
3
|
Lefèvre G, Gibier JB, Bongiovanni A, Lhermitte L, Rossignol J, Anglo E, Dendooven A, Dubois R, Terriou L, Launay D, Barete S, Esnault S, Frenzel L, Gourguechon C, Ballul T, Dezoteux F, Staumont-Salle D, Copin MC, Rignault-Bricard R, Maciel TT, Damaj G, Tardivel M, Crinquette-Verhasselt M, Dubreuil P, Maouche-Chrétien L, Bruneau J, Lortholary O, Duployez N, Behal H, Molina TJ, Hermine O. Interactions between eosinophils and IL-5Rα-positive mast cells in nonadvanced systemic mastocytosis. J Allergy Clin Immunol 2024; 154:1523-1533. [PMID: 39151478 DOI: 10.1016/j.jaci.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/27/2024] [Accepted: 07/10/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Bidirectional interactions between eosinophils and mast cells (MCs) have been reported in various allergic diseases. Bone marrow (BM) eosinophilia, and to a lesser extent blood eosinophilia, is common in systemic mastocytosis (SM), but its significance remains unknown. OBJECTIVE We described blood and BM eosinophil characteristics in SM. METHODS A large collection of BM biopsy samples was analyzed using immunohistochemical staining and whole-slide imaging. Eosinophil and extracellular granules were detected by eosinophil peroxidase (EPX) staining and MCs by KIT staining. Complementary analyses were conducted using flow cytometry and immunofluorescence. RESULTS Eosinophil infiltrates and large areas of eosinophil degranulation were observed within or around BM MC infiltrates in SM. EPX staining surface, highlighting intact eosinophils and eosinophil degranulation, was higher in nonadvanced SM (n = 37 BM biopsy samples) compared with both controls (n = 8, P = .0003) and advanced SM (n = 24, P = .014). In nonadvanced SM, positive correlations were observed between serum tryptase levels and percentages of eosinophil counts in BM aspirations (Spearman r coefficient r = 0.38, P = .038), eosinophils count in BM biopsy samples (r = 0.45, P = .007), EPX staining (r = 0.37, P = .035), and eosinophil degranulation (r = 0.39, P = .023). Eosinophil counts in BM biopsy samples also correlated with MC counts (r = 0.47, P = .006) and KIT staining surface (r = 0.49, P = .003). BM MCs expressed IL-5 receptor and other usual eosinophil cytokine/chemokine receptors, and blood eosinophils displayed several increased surface markers compared with controls, suggesting an activated state. CONCLUSION Our data suggest possible cross talk between MCs and eosinophils, supporting MC tryptase release and MC activation-related symptoms. This suggests a rationale for targeting eosinophils in nonadvanced SM not fully controlled by other therapies.
Collapse
Affiliation(s)
- Guillaume Lefèvre
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO).
| | - Jean-Baptiste Gibier
- University of Lille, Institut de Pathologie, Centre de Biopathologie, CHU Lille, Lille, France
| | - Antonino Bongiovanni
- Centre National de la Recherche Scientifique (CNRS), INSERM, CHU Lille, University of Lille, Institut Pasteur de Lille, Lille, France
| | - Ludovic Lhermitte
- Laboratoire d'Onco-Hématologie, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Julien Rossignol
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Emilie Anglo
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France
| | - Arnaud Dendooven
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France
| | - Romain Dubois
- University of Lille, Institut de Pathologie, Centre de Biopathologie, CHU Lille, Lille, France
| | - Louis Terriou
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Département de Médecine Interne et Immunologie Clinique, CHU Lille, Lille, France
| | - David Launay
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Département de Médecine Interne et Immunologie Clinique, CHU Lille, Lille, France
| | - Stéphane Barete
- CEREMAST, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Unit de Dermatologie, Sorbonne Université Paris, Paris, France
| | - Stéphane Esnault
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; Institut d'Immunologie, CHU Lille, Lille, France; Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wis
| | - Laurent Frenzel
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | | | - Thomas Ballul
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Frédéric Dezoteux
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Department of Dermatology, CHU Lille, Lille, France
| | - Delphine Staumont-Salle
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire (CHU) Lille, and Institute for Translational Research in Inflammation (INFINITE), Lille, France; National Reference Center for Hypereosinophilic Syndromes (CEREO); Department of Dermatology, CHU Lille, Lille, France
| | - Marie-Christine Copin
- Department of Pathology, CHU Angers, University of Angers, INSERM, CNRS, CRCI(2)NA, Angers, France
| | | | | | - Gandhi Damaj
- Institut d'Hématologie, University of Caen Normandie, Caen, France
| | - Meryem Tardivel
- Centre National de la Recherche Scientifique (CNRS), INSERM, CHU Lille, University of Lille, Institut Pasteur de Lille, Lille, France
| | | | - Patrice Dubreuil
- Signaling, Hematopoiesis, and Mechanism of Oncogenesis (CRCM), CEREMAST, and Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytose (AFIRMM) studies, INSERM U1068; Institut Paoli-Calmettes; UM105, Aix-Marseille University; and CNRS, UMR7258, Marseille, France
| | | | - Julie Bruneau
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Olivier Lortholary
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Nicolas Duployez
- CNRS, INSERM, CHU Lille, Cancer Heterogeneity, Plasticity, and Resistance to Therapies (CANTHER), University of Lille, Institut d'Hématologie, CHU Lille, Lille, France
| | - Hélène Behal
- University of Lille, CHU Lille, Evaluation des Technologies de Santé et des Pratiques Médicales (METRICS), Lille, France
| | - Thierry Jo Molina
- University of Paris, Institut Imagine, INSERM, Paris, France; Signaling, Hematopoiesis, and Mechanism of Oncogenesis (CRCM), CEREMAST, and Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytose (AFIRMM) studies, INSERM U1068; Institut Paoli-Calmettes; UM105, Aix-Marseille University; and CNRS, UMR7258, Marseille, France
| | - Olivier Hermine
- University of Paris, Institut Imagine, INSERM, Paris, France; French Reference Center for Mastocytosis (CEREMAST), Hôpital Necker-Enfants Malades, AP-HP, Paris, France; Department of Hematology, Hôpital Necker-Enfants Malades, AP-HP, Paris, France.
| |
Collapse
|
4
|
Takla A, Shah P, Sbenghe M, Baibhav B, Feitell S. Incessant Ventricular Tachycardia: An Atypical Presentation of Chronic Eosinophilic Leukemia. JACC Case Rep 2024; 29:102461. [PMID: 39295803 PMCID: PMC11405962 DOI: 10.1016/j.jaccas.2024.102461] [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: 02/18/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 09/21/2024]
Abstract
Hypereosinophilic syndrome comprises a diverse and intricate array of rare disorders, exhibiting clinical manifestations that extend across various medical subspecialties. Within its myeloid form, chronic eosinophilic leukemia represents a rare myeloid malignancy characterized by severe hematological complications and distinctive organ dysfunction, notably affecting the cardiovascular system. This report presents a rare case of chronic eosinophilic leukemia and Loeffler syndrome with an initial presentation of ventricular tachycardia.
Collapse
Affiliation(s)
- Andrew Takla
- Department of Internal Medicine, Rochester General Hospital, Rochester, New York, USA
| | - Purva Shah
- Department of Internal Medicine, Rochester General Hospital, Rochester, New York, USA
| | - Maria Sbenghe
- Department of Hematology and Oncology, Rochester General Hospital, Rochester, New York, USA
| | - Bipul Baibhav
- Sands Constellation Heart Institute, Rochester Regional Health, Rochester, New York, USA
| | - Scott Feitell
- Sands Constellation Heart Institute, Rochester Regional Health, Rochester, New York, USA
| |
Collapse
|
5
|
Shomali W, Gotlib J. World Health Organization and International Consensus Classification of eosinophilic disorders: 2024 update on diagnosis, risk stratification, and management. Am J Hematol 2024; 99:946-968. [PMID: 38551368 DOI: 10.1002/ajh.27287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 04/09/2024]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of non-hematologic (secondary or reactive) and hematologic (primary or clonal) disorders with the potential for end-organ damage. DIAGNOSIS Hypereosinophilia (HE) has generally been defined as a peripheral blood eosinophil count greater than 1.5 × 109/L, and may be associated with tissue damage. After the exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of various tests. They include morphologic review of the blood and marrow, standard cytogenetics, fluorescence in situ hybridization, molecular testing and flow immunophenotyping to detect histopathologic or clonal evidence for an acute or chronic hematolymphoid neoplasm. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2022 World Health Organization and International Consensus Classification endorse a semi-molecular classification scheme of disease subtypes. This includes the major category "myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions" (MLN-eo-TK), and the MPN subtype, "chronic eosinophilic leukemia" (CEL). Lymphocyte-variant HE is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g., <1.5 × 109/L) without symptoms or signs of organ involvement, a watch and wait approach with close follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Pemigatinib was recently approved for patients with relapsed or refractory FGFR1-rearranged neoplasms. Corticosteroids are first-line therapy for patients with lymphocyte-variant HE and HES. Hydroxyurea and interferon-α have demonstrated efficacy as initial treatment and in steroid-refractory cases of HES. Mepolizumab, an interleukin-5 (IL-5) antagonist monoclonal antibody, is approved by the U.S Food and Drug Administration for patients with idiopathic HES. Cytotoxic chemotherapy agents, and hematopoietic stem cell transplantation have been used for aggressive forms of HES and CEL, with outcomes reported for limited numbers of patients. Targeted therapies such as the IL-5 receptor antibody benralizumab, IL-5 monoclonal antibody depemokimab, and various tyrosine kinase inhibitors for MLN-eo-TK, are under active investigation.
Collapse
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California, USA
| | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Abstract
Systemic mastocytosis is associated with KIT D816V mutation in more than 90% of cases. Patients with non-advanced forms of mastocytosis (indolent systemic mastocytosis, bone marrow mastocytosis, and smoldering systenic mastocytosis) have a low rate of progession to advanced variants and generally have a comparable life expectancy to age-matched general population. Symptomatology in non-advanced mastocytosis is variable and is related to mast cell mediator release. While some patients require no or minimal symptomatic therapy with antimediator drugs, other may suffer from refractory symptoms impacting the quality of life despite being on multiple anti-mediator drugs. KIT tyrosine kinase inhibitors have been approved for advanced SM, and avapritinib has also been recently approved as the first such inhibitor for indolent systemic mastocytosis. Other TKIs are currently in clinical trials for patients with non-advanced SM who have persistent and severe symptoms despite optimized antimediator therapy. This article will review the current state of the science and available clinical data from trials of tyrosine kinase inhibitors in non-advanced systemic mastocytosis.
Collapse
Affiliation(s)
- Cem Akin
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan, 24 Frank Lloyd Wright Drive, PO Box 442, Suite H-2100, Ann Arbor, MI 48106-0442, USA.
| |
Collapse
|
7
|
Pardanani A. Systemic mastocytosis in adults: 2023 update on diagnosis, risk stratification and management. Am J Hematol 2023; 98:1097-1116. [PMID: 37309222 DOI: 10.1002/ajh.26962] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 06/14/2023]
Abstract
OVERVIEW Systemic mastocytosis (SM) results from clonal proliferation of mast cells (MC) in extracutaneous organs. DIAGNOSIS The major criterion is presence of multifocal MC clusters in the bone marrow and/or extracutaneous organs. Minor diagnostic criteria include elevated serum tryptase level, MC CD25/CD2/CD30 expression, and presence of activating KIT mutations. RISK STRATIFICATION Establishing SM subtype as per the International Consensus Classification/World Health Organization classification systems is an important first step. Patients either have indolent/smoldering SM (ISM/SSM) or advanced SM, including aggressive SM (ASM), SM with associated myeloid neoplasm (SM-AMN), and mast cell leukemia. Identification of poor-risk mutations (i.e., ASXL1, RUNX1, SRSF2, NRAS) further refines the risk stratification. Several risk models are available to help assign prognosis in SM patients. MANAGEMENT Treatment goals for ISM patients are primarily directed toward anaphylaxis prevention/symptom control/osteoporosis treatment. Patients with advanced SM frequently need MC cytoreductive therapy to reverse disease-related organ dysfunction. Tyrosine kinase inhibitors (TKI) (midostaurin, avapritinib) have changed the treatment landscape in SM. While deep biochemical, histological and molecular responses have been documented with avapritinib treatment, its efficacy as monotherapy against a multimutated AMN disease component in SM-AMN patients remains unclear. Cladribine continues to have a role for MC debulking, whereas interferon-α has a diminishing role in the TKI era. Treatment of SM-AMN primarily targets the AMN component, particularly if an aggressive disease such as acute leukemia is present. Allogeneic stem cell transplant has a role in such patients. Imatinib has a therapeutic role only in the rare patient with an imatinib-sensitive KIT mutation.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Groh M, Rohmer J, Etienne N, Abou Chahla W, Baudet A, Chan Hew Wai A, Chenivesse C, Clisson Rusek I, Cottin V, Decamp M, De Groote P, Delahousse F, Duployez N, Faguer S, Gottrand F, Huang F, Leblanc T, Magnan A, Martin T, Mortuaire G, Néel A, Paris L, Petit A, Rossignol J, Schleinitz N, Soret-Dulphy J, Staumont-Salle D, Terrier B, Terriou L, Viallard JF, Lefèvre G, Kahn JE. French guidelines for the etiological workup of eosinophilia and the management of hypereosinophilic syndromes. Orphanet J Rare Dis 2023; 18:100. [PMID: 37122022 PMCID: PMC10148979 DOI: 10.1186/s13023-023-02696-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/02/2023] [Indexed: 05/02/2023] Open
Abstract
Eosinophilic-related clinical manifestations are protean and the underlying conditions underpinning eosinophilia are highly diverse. The etiological workup of unexplained eosinophilia/hypereosinophilia can be challenging, and can lead sometimes to extensive, inappropriate, costly and/or invasive investigations. To date, guidelines for the etiological workup and management of eosinophilia are mainly issued by hematologists, and thus mostly cover the scope of clonal hypereosinophilic syndromes (HES). Here, thanks to an extensive literature review, and thanks to the joint work of a large panel of experts involving physicians from both adult and pediatric medicine and from various subspecialties (as well as a representative of a patients' association representative), we provide recommendations for both the step-by step diagnostic workup of eosinophilia (whether unexplained or within specific contexts) as well as the management and follow-up of the full spectrum of eosinophilic disorders (including clonal, reactive, lymphocytic and idiopathic HES, as well as single-organ diseases). Didactic prescription summaries intended to facilitate the prescription of eosinophil-targeted drugs are also provided, as are practical diagnostic and therapeutic algorithms. Lastly, this set of recommendations also includes a summary intended for general practitioners, as well as an overview of the therapeutic patient education program set up by the French reference center for HES. Further updates will be mandatory as new validated information emerges.
Collapse
Affiliation(s)
- Matthieu Groh
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France.
- Department of Internal Medicine, Hôpital Foch, Suresnes, France.
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France.
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France.
| | - Julien Rohmer
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, University of Sorbonne-Paris-Cité, APHP, CHU Bichat, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Nicolas Etienne
- Department of Infectious Diseases and Tropical Medicine, University of Sorbonne-Paris-Cité, APHP, CHU Necker-Enfants Malades, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Wadih Abou Chahla
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Pediatric Hematology, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Antoine Baudet
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CH Annecy Genevois, Metz Tessy, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Aurélie Chan Hew Wai
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Pharmacology, Hôpital Foch, Suresnes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Cécile Chenivesse
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- CNRS, Inserm, Institut Pasteur de Lille, U1019-UMR9017-CIIL-Centre d'Infection et d'Immunité de Lille, University of Lille, CHU Lille, Lille, France
- CRISALIS (Clinical Research Initiative in Severe Asthma: a Lever for Innovation and Science), F-CRIN Network, INSERM US015, Toulouse, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Irena Clisson Rusek
- Association Pour l'Information sur les Maladies à Eosinophiles, Bourg-la-Reine, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Vincent Cottin
- Department of Respiratory Medicine, Hôpital Louis Pradel, UMR754 INRAE, University of Lyon 1, Hospices Civils de Lyon, Lyon, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Matthieu Decamp
- Department of Cytogenetics, CHU de Caen, Caen, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Pascal De Groote
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Cardiology, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Fanny Delahousse
- , Nantes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Nicolas Duployez
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Laboratory of Hematology, University of Lille, CHU Lille, Lille, France
- CNRS, Inserm, IRCL, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Stanislas Faguer
- Department of Nephrology and Organ Transplantation, University of Paul Sabatier Toulouse III, CHU Toulouse, Toulouse, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Frédéric Gottrand
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
- Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Jeanne de Flandre Children's Hospital, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Florent Huang
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Cardiology, Hôpital Foch, Suresnes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Thierry Leblanc
- Pediatric Hematology and Immunology Department, University Sorbonne-Paris-Cité, APHP, CHU Robert Debré, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Antoine Magnan
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Respiratory Medicine, Hôpital Foch, Suresnes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Thierry Martin
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CHU Strasbourg, Strasbourg, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Geoffrey Mortuaire
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
- Otorhinolaryngology-Head and Neck Department, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Antoine Néel
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CHU Nantes, Nantes, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Luc Paris
- Department of Parasitology and Mycology, Sorbonne Université, APHP, CHU Pitié-Salpêtrière, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Arnaud Petit
- Department of Hematology and Pediatric Oncology, Sorbonne Université, APHP, CHU Armand Trousseau, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Julien Rossignol
- Department of Hematology, University of Sorbonne-Paris-Cité, APHP, CHU Necker, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Nicolas Schleinitz
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, APHM, CHU La Timone, Marseille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Juliette Soret-Dulphy
- Centre d'Investigation Clinique, University of Sorbonne-Paris-Cité, AP-HP, CHU St-Louis, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Delphine Staumont-Salle
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Dermatology, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Benjamin Terrier
- Department of Internal Medicine, University of Sorbonne-Paris-Cité, AP-HP, Paris, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Louis Terriou
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine and Clinical Immunology, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Jean-François Viallard
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Department of Internal Medicine, CHU de Bordeaux, Bordeaux, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Guillaume Lefèvre
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Inserm, U1286 - INFINITE - Institute for Translational Research in Inflammation, University of Lille, CHU Lille, Lille, France
- Department of Internal Medicine and Clinical Immunology, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, CHU de Bordeaux, Bordeaux, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| | - Jean-Emmanuel Kahn
- Department of Internal Medicine, National Reference Center for Hypereosinophilic Syndromes (CEREO), Hôpital Foch, 40, Rue Worth, 92151, Suresnes, France
- Institut d'Immunologie, University of Lille, CHU de Lille, Lille, France
- Department of Internal Medicine, University of Paris Saclay, APHP, CHU Ambroise Paré, Boulogne-Billancourt, France
| |
Collapse
|
9
|
Chronic Eosinophilic Leukemia Presenting as Cardiac Failure. Case Rep Hematol 2022; 2022:7841310. [PMID: 36568337 PMCID: PMC9788895 DOI: 10.1155/2022/7841310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/13/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Chronic eosinophilic leukemia (CEL) is a rare chronic myeloproliferative disorder characterized by sustained eosinophilia. Although the incidence of CEL is uncertain, it can be clinically devastating as it has a propensity to affect several important organ systems. This is of particular significance in Sub-Saharan Africa where helminthic infections are a more prevalent cause of eosinophilia. To the best of our knowledge, we present the first reported case of CEL complicated by cardiac disease in a Ghanaian. He presented with a history of orthopnoea and dyspnoea on exertion, and examination revealed a pansystolic murmur over the mitral region and moderate splenomegaly. Good symptomatic control was achieved using hydroxyurea after which haematologic and cytogenetic remission was achieved after 12 weeks on a tyrosine kinase inhibitor. Physicians working in low resource environments should exclude clonality in patients presenting with eosinophilia and end-organ damage.
Collapse
|
10
|
Gotlib J. Available and emerging therapies for bona fide advanced systemic mastocytosis and primary eosinophilic neoplasms. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:34-46. [PMID: 36485158 PMCID: PMC9821059 DOI: 10.1182/hematology.2022000368] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The historically poor prognosis of patients with advanced systemic mastocytosis (AdvSM) and primary eosinophilic neoplasms has shifted to increasingly favorable outcomes with the discovery of druggable targets. The multikinase/KIT inhibitor midostaurin and the highly selective KIT D816V inhibitor avapritinib can elicit marked improvements in measures of mast cell (MC) burden as well as reversion of MC-mediated organ damage (C-findings) and disease symptoms. With avapritinib, the achievement of molecular remission of KIT D816V and improved survival compared with historical therapy suggests a potential to affect disease natural history. BLU-263 and bezuclastinib are KIT D816V inhibitors currently being tested in trials of AdvSM. In the new World Health Organization and International Consensus Classifications, the category of "myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase (TK) gene fusions" is inclusive of rearrangements involving PDGFRA, PDGFRB, FGFR1, JAK2, FLT3, and ETV6::ABL1. While the successful outcomes with imatinib in FIP1L1::PDGFRA-positive cases and PDGFRB-rearranged neoplasms have become the "poster children" of these disorders, the responses of the other TK-driven neoplasms to small-molecule inhibitors are more variable. The selective FGFR inhibitor pemigatinib, approved in August 2022, is a promising therapy in aggressive FGFR1-driven diseases and highlights the role of such agents in bridging patients to allogeneic transplantation. This review summarizes the data for these approved and investigational agents and discusses open questions and future priorities regarding the management of these rare diseases.
Collapse
Affiliation(s)
- Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
11
|
Velloso EDRP, Padulla GA, de Cerqueira AMM, de Sousa AM, Sandes AF, Traina F, Seguro FS, Nogueira FL, Pereira GDF, Boechat JL, Pagnano KBB, Marchi LL, Ensina LF, Giavina-Bianchi M, Aun MV, Agondi RC, Santos FPDS, Giavina-Bianchi P. Diagnosis and treatment of systemic mastocytosis in Brazil: Recommendations of a multidisciplinary expert panel. Hematol Transfus Cell Ther 2022; 44:582-594. [PMID: 35688791 PMCID: PMC9605912 DOI: 10.1016/j.htct.2022.04.006] [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: 12/10/2021] [Revised: 03/04/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Systemic Mastocytosis comprises a group of neoplastic diseases characterized by clonal expansion and infiltration of mast cells into several organs. The diagnosis and treatment of this disease may be challenging for non-specialists. OBJECTIVE Make suggestions or recommendations in Systemic Mastocytosis based in a panel of Brazilian specialists. METHOD AND RESULTS An online expert panel with 18 multidisciplinary specialists was convened to propose recommendations on the diagnosis and treatment of Systemic Mastocytosis in Brazil. Recommendations were based on discussions of topics and multiple-choice questions and were graded using the Oxford Centre for Evidence-Based Medicine 2011 Levels of Evidence Chart. CONCLUSION Twenty-two recommendations or suggestions were proposed based on a literature review and graded according to the findings.
Collapse
Affiliation(s)
- Elvira D Rodrigues Pereira Velloso
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Geórgia A Padulla
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Adriana Martins de Sousa
- Instituto de Pediatria e Puericultura Martagão Gesteira da Universidade Federal do Rio de Janeiro (IPPMG UFRJ), Rio de Janeiro, RJ, Brazil
| | - Alex Freire Sandes
- Grupo Fleury, São Paulo, SP, Brazil; Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brazil
| | - Fabiola Traina
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (FMRP USP), São Paulo, SP, Brazil
| | - Fernanda Salles Seguro
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Frederico Lisboa Nogueira
- Hospital Luxemburgo, Instituto Mário Penna, Belo Horizonte, MG, Brazil; Grupo Oncoclínicas, Belo Horizonte, MG, Brazil
| | | | - José Laerte Boechat
- Faculdade de Medicina da Universidade Federal Fluminense (FM UFF), Niteroi, RJ, Brazil
| | | | - Luan Lima Marchi
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Mara Giavina-Bianchi
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marcelo Vivolo Aun
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil; Faculdade Israelita de Ciências da Saúde Albert Einstein, São Paulo, SP, Brazil
| | - Rosana Câmara Agondi
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Fabio Pires de Souza Santos
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil; A Beneficência Portuguesa de São Paulo, BP, São Paulo, SP, Brazil
| | - Pedro Giavina-Bianchi
- Clinical Immunology and Allergy Division. Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
12
|
Arber DA, Orazi A, Hasserjian RP, Borowitz MJ, Calvo KR, Kvasnicka HM, Wang SA, Bagg A, Barbui T, Branford S, Bueso-Ramos CE, Cortes JE, Dal Cin P, DiNardo CD, Dombret H, Duncavage EJ, Ebert BL, Estey EH, Facchetti F, Foucar K, Gangat N, Gianelli U, Godley LA, Gökbuget N, Gotlib J, Hellström-Lindberg E, Hobbs GS, Hoffman R, Jabbour EJ, Kiladjian JJ, Larson RA, Le Beau MM, Loh MLC, Löwenberg B, Macintyre E, Malcovati L, Mullighan CG, Niemeyer C, Odenike OM, Ogawa S, Orfao A, Papaemmanuil E, Passamonti F, Porkka K, Pui CH, Radich JP, Reiter A, Rozman M, Rudelius M, Savona MR, Schiffer CA, Schmitt-Graeff A, Shimamura A, Sierra J, Stock WA, Stone RM, Tallman MS, Thiele J, Tien HF, Tzankov A, Vannucchi AM, Vyas P, Wei AH, Weinberg OK, Wierzbowska A, Cazzola M, Döhner H, Tefferi A. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data. Blood 2022; 140:1200-1228. [PMID: 35767897 PMCID: PMC9479031 DOI: 10.1182/blood.2022015850] [Citation(s) in RCA: 1350] [Impact Index Per Article: 450.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/16/2022] [Indexed: 02/02/2023] Open
Abstract
The classification of myeloid neoplasms and acute leukemias was last updated in 2016 within a collaboration between the World Health Organization (WHO), the Society for Hematopathology, and the European Association for Haematopathology. This collaboration was primarily based on input from a clinical advisory committees (CACs) composed of pathologists, hematologists, oncologists, geneticists, and bioinformaticians from around the world. The recent advances in our understanding of the biology of hematologic malignancies, the experience with the use of the 2016 WHO classification in clinical practice, and the results of clinical trials have indicated the need for further revising and updating the classification. As a continuation of this CAC-based process, the authors, a group with expertise in the clinical, pathologic, and genetic aspects of these disorders, developed the International Consensus Classification (ICC) of myeloid neoplasms and acute leukemias. Using a multiparameter approach, the main objective of the consensus process was the definition of real disease entities, including the introduction of new entities and refined criteria for existing diagnostic categories, based on accumulated data. The ICC is aimed at facilitating diagnosis and prognostication of these neoplasms, improving treatment of affected patients, and allowing the design of innovative clinical trials.
Collapse
Affiliation(s)
| | - Attilio Orazi
- Texas Tech University Health Sciences Center El Paso, El Paso, TX
| | | | | | | | | | - Sa A Wang
- University of Texas MD Anderson Cancer Center, Houston, TX
| | - Adam Bagg
- University of Pennsylvania, Philadelphia, PA
| | - Tiziano Barbui
- Clinical Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | | | | | | | | | | | - Hervé Dombret
- Université Paris Cité, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, Paris, France
| | | | | | | | | | | | | | | | | | | | - Jason Gotlib
- Stanford University School of Medicine, Stanford, CA
| | | | | | | | | | - Jean-Jacques Kiladjian
- Université Paris Cité, Hôpital Saint-Louis, Assistance Publique - Hôpitaux de Paris, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kimmo Porkka
- Helsinki University Central Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | | | | | | | | | | | | | | | - Akiko Shimamura
- Dana-Farber Cancer Institute, Boston, MA
- Boston Children's Cancer and Blood Disorders Center, Boston, MA
| | - Jorge Sierra
- Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | | - Hwei-Fang Tien
- National Taiwan University Hospital, Taipei City, Taiwan
| | | | | | - Paresh Vyas
- University of Oxford, Oxford, United Kingdom
| | - Andrew H Wei
- Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
13
|
Shomali W, Gotlib J. World Health Organization-defined eosinophilic disorders: 2022 update on diagnosis, risk stratification, and management. Am J Hematol 2022; 97:129-148. [PMID: 34533850 DOI: 10.1002/ajh.26352] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/13/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of nonhematologic (secondary or reactive) and hematologic (primary or clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia (HE) has generally been defined as a peripheral blood eosinophil count greater than 1.5 × 109 /L. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on morphologic review of the blood and marrow, standard cytogenetics, fluorescence in situ hybridization, next generation sequencing gene assays, and flow immunophenotyping to detect histopathologic or clonal evidence for an acute or chronic hematolymphoid neoplasm. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2016 World Health Organization endorses a semi-molecular classification scheme of disease subtypes. This includes the major category "myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1 or with PCM1-JAK2", and the myeloproliferative neoplasm subtype, "chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS). Lymphocyte-variant HE is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (eg, < 1.5 × 109 /L) without symptoms or signs of organ involvement, a watch and wait approach with close follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant HE and HES. Hydroxyurea and interferon-α have demonstrated efficacy as initial treatment and in steroid-refractory cases of HES. Mepolizumab, an interleukin-5 (IL-5) antagonist monoclonal antibody, was recently approved by the US Food and Drug Administration for patients with idiopathic HES. The use of the IL-5 receptor antibody benralizumab, as well as other targeted therapies such as JAK2 and FGFR1 inhibitors, is under active investigation.
Collapse
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute Stanford University School of Medicine Stanford California USA
| | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute Stanford University School of Medicine Stanford California USA
| |
Collapse
|
14
|
Pardanani A. Systemic mastocytosis in adults: 2021 Update on diagnosis, risk stratification and management. Am J Hematol 2021; 96:508-525. [PMID: 33524167 DOI: 10.1002/ajh.26118] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
OVERVIEW Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in extra-cutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of spindled MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC CD25 expression, and presence of KITD816V mutation. RISK STRATIFICATION Establishing SM subtype as per the World Health Organization classification system is an important first step. Broadly, patients either have indolent/smoldering SM (ISM/SSM) or advanced SM, the latter includes aggressive SM (ASM), SM with associated hematological neoplasm (SM-AHN), and mast cell leukemia (MCL). Identification of poor-risk mutations (ie, ASXL1, RUNX1, SRSF2, NRAS) further refines the risk stratification. Recently, clinical and hybrid clinical-molecular risk models have been developed to more accurately assign prognosis in SM patients. MANAGEMENT Treatment goals for ISM patients are primarily directed towards anaphylaxis prevention/symptom control/osteoporosis treatment. Patients with advanced SM frequently need MC cytoreductive therapy to ameliorate disease-related organ dysfunction. High response rates have been seen with small-molecule inhibitors that target mutant-KIT, including midostaurin (Food and Drug Administration approved) or avapritinib (investigational). Other options for MC cytoreduction include cladribine or interferon-α, although head-to-head comparisons are lacking. Treatment of SM-AHN primarily targets the AHN component, particularly if an aggressive disease such as acute myeloid leukemia is present. Allogeneic stem cell transplant can be considered in such patients, or in those with relapsed/refractory advanced SM. Imatinib has a limited therapeutic role in SM; effective cytoreduction is limited to those with imatinib-sensitive KIT mutations.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology, Department of Medicine Mayo Clinic Rochester Minnesota
| |
Collapse
|
15
|
Gerds AT, Gotlib J, Bose P, Deininger MW, Dunbar A, Elshoury A, George TI, Gojo I, Gundabolu K, Hexner E, Hobbs G, Jain T, Jamieson C, Kuykendall AT, McMahon B, Mohan SR, Oehler V, Oh S, Pardanani A, Podoltsev N, Ranheim E, Rein L, Salit R, Snyder DS, Stein BL, Talpaz M, Thota S, Vachhani P, Wadleigh M, Walsh K, Ward DC, Bergman MA, Sundar H. Myeloid/Lymphoid Neoplasms with Eosinophilia and TK Fusion Genes, Version 3.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2020; 18:1248-1269. [PMID: 32886902 DOI: 10.6004/jnccn.2020.0042] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Eosinophilic disorders and related syndromes represent a heterogeneous group of neoplastic and nonneoplastic conditions, characterized by more eosinophils in the peripheral blood, and may involve eosinophil-induced organ damage. In the WHO classification of myeloid and lymphoid neoplasms, eosinophilic disorders characterized by dysregulated tyrosine kinase (TK) fusion genes are recognized as a new category termed, myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB or FGFR1 or with PCM1-JAK2. In addition to these aforementioned TK fusion genes, rearrangements involving FLT3 and ABL1 genes have also been described. These new NCCN Guidelines include recommendations for the diagnosis, staging, and treatment of any one of the myeloid/lymphoid neoplasms with eosinophilia (MLN-Eo) and a TK fusion gene included in the 2017 WHO Classification, as well as MLN-Eo and a FLT3 or ABL1 rearrangement.
Collapse
Affiliation(s)
- Aaron T Gerds
- Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | | | | | | | - Ivana Gojo
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | - Tania Jain
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | - Vivian Oehler
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Stephen Oh
- Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | | | | | | | | | - Rachel Salit
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Brady L Stein
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | - Katherine Walsh
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Dawn C Ward
- UCLA Jonsson Comprehensive Cancer Center; and
| | | | | |
Collapse
|
16
|
Sreedharanunni S, Sachdeva MUS, Sharma S, Rana S, Sandal R, Kumar N, Sharma P, Naseem S, Ahluwalia J, Das R, Malhotra P, Varma N. Paratrabecular myelofibrosis and occult mastocytosis are strong morphological clues to suspect FIP1L1-PDGFRA translocation in hypereosinophilia. Indian J Hematol Blood Transfus 2020; 36:384-389. [PMID: 32425395 DOI: 10.1007/s12288-019-01236-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/22/2019] [Indexed: 11/25/2022] Open
Abstract
To study the clinico-haematological and histopathological characteristics of FIP1L1-PDGFRA rearranged hypereosinophilia/hypereosinophilic syndrome (F/P+ve HE/HES), a retrospective analysis of patients with F/P+ve HE diagnosed over a period of 43 months was performed. Peripheral blood smears, bone marrow aspirate (BMA) and biopsies (BMB) were reviewed in each case and; reticulin stain and immunohistochemistry for mast cell tryptase (MCT) and CD117 was performed. F/P+ve HE was diagnosed in a total of ten patients during study period. All patients were males with a median age of 36 years (23-44 years). The median duration of presenting complaints was 7 months (2 months-3 years) which included specific symptoms related to various organs (80% of cases). Anaemia, thrombocytopenia and splenomegaly were seen in 60%, 50% and 90% of the cases respectively. Mastocytosis was not obvious in BMA but identified by MCT on BMB in all cases. Myelofibrosis (grade ≥ 1) was seen in 80% of the cases and includes multifocal paratrabecular fibrosis in 50% of the biopsies. Our study shows that bone marrow mastocytosis and myelofibrosis are very useful morphological indicators to suspect F/P+ve HE and suggests the routine use of reticulin staining and MCT immunohistochemistry in all BMBs performed for the evaluation of HE/HES.
Collapse
Affiliation(s)
- Sreejesh Sreedharanunni
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Man Updesh Singh Sachdeva
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Saniya Sharma
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Sonia Rana
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Rajeev Sandal
- 2Adult Clinical Hematology and Bone Marrow Transplantation Unit, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012 India
| | - Narender Kumar
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Prashant Sharma
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Shano Naseem
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Jasmina Ahluwalia
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Reena Das
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| | - Pankaj Malhotra
- 2Adult Clinical Hematology and Bone Marrow Transplantation Unit, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012 India
| | - Neelam Varma
- 1Department of Hematology, Post Graduate Institute of Medical Education and Research, 5th floor, Research block A, Chandigarh, 160012 India
| |
Collapse
|
17
|
Yang Y, Lin H, Du Z, Hu R, Tang Y, Liang X, Sun J, Tan Y. Imatinib therapy in acute myeloid leukemia with DEK-NUP214 and FIP1L1-PDGFRA rearrangement: A case report. Oncol Lett 2020; 19:3587-3592. [PMID: 32269633 DOI: 10.3892/ol.2020.11455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 01/07/2020] [Indexed: 11/06/2022] Open
Abstract
The fusion product of FIP1-like-1 (FIP1L1) and platelet-derived growth factor receptor α (PDGFRA) gene rearrangement is a tyrosine kinase oncoprotein sensitive to imatinib. This gene rearrangement characterizes a novel clinico-biological class of myeloid and lymphoid neoplasms with eosinophilia and PDGFRA abnormalities. The DEK proto-oncogene (DEK) and nucleoporin 214 (NUP214) rearrangement is rare in patients with acute myeloid leukemia (AML); therefore, the coexistence of DEK-NUP214 and FIP1L1-PDGFRA rearrangements in patients with AML is extremely rare. The present study presents a rare relapse case of a patient with AML with DEK-NUP214 and FIP1L1-PDGFRA rearrangements, without marked eosinophilia in the peripheral blood or bone marrow. Low-dose imatinib monotherapy without intensive chemotherapy was used to achieve complete hematological remission.
Collapse
Affiliation(s)
- Yanping Yang
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hai Lin
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhonghua Du
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruiping Hu
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Tang
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xinyue Liang
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jingnan Sun
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yehui Tan
- Department of Hematology, First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
18
|
Moalong KMC, Diestro JDB, Espiritu AI, Lucero JAC, de la Cruz APRL, Valencia JM, Chua EM, Cenina ARF, Spears J, San Jose MCZ. FIP1L1-PDGFRA-Positive Hypereosinophilia Presenting with Bilateral Extracranial Carotid Artery Aneurysms. World Neurosurg 2020; 139:419-422. [PMID: 32360918 DOI: 10.1016/j.wneu.2020.04.119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To describe a case of an adult female Filipino with hypereosinophilia and bilateral carotid artery aneurysms who subsequently developed bilateral cerebral hemisphere strokes following aneurysm stenting. CASE DESCRIPTION A 57-year-old female patient with persistent hypereosinophilia presented with progressively enlarging bilateral neck masses, revealed to be carotid artery aneurysms on computed tomography angiography. Following surgical exploration, she later developed right-sided hemiplegia, aphasia, and right hemianopia. Cranial computed tomography revealed infarcts on both middle cerebral artery territories. Bone marrow biopsy and fluorescent in situ hybridization revealed findings suggestive of hypereosinophilic syndrome. She was started on standard aspirin and statin therapy and was discharged sixteen days after the procedure. Partial improvement of neurologic deficits was noted two months later on follow up. Chemotherapy with imatinib was initiated. CONCLUSIONS This patient's prothrombotic state from FIP1L1-PDGFRA-positive hypereosinophilia may have led to large carotid artery aneurysm formation and intramural thrombosis. This case demonstrates a possible and heretofore undocumented neurovascular sequela of hypereosinophilic syndrome.
Collapse
Affiliation(s)
- Kevin Michael Cachapero Moalong
- Division of Adult Neurology, Department of Neurosciences, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines.
| | - Jose Danilo Bengzon Diestro
- Division of Diagnostic and Therapeutic Neuroradiology, Department of Medical Imaging, St. Michael's Hospital, University of Toronto, Toronto, Canada
| | - Adrian Isidoro Espiritu
- Division of Adult Neurology, Department of Neurosciences, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines; Department of Clinical Epidemiology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Josephine Anne Cuevas Lucero
- Department of Medicine, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines; Division of Hematology, Department of Medicine, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | | | - Jeffrey Matubis Valencia
- Department of Medicine, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines; Division of Cardiology, Department of Medicine, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Enrique Melarin Chua
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Alvin Rae Francisco Cenina
- Division of Adult Neurology, Department of Neurosciences, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| | - Julian Spears
- Division of Diagnostic and Therapeutic Neuroradiology, Department of Medical Imaging, St. Michael's Hospital, University of Toronto, Toronto, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - Maria Cristina Zarsadias San Jose
- Division of Adult Neurology, Department of Neurosciences, College of Medicine and Philippine General Hospital, University of the Philippines Manila, Manila, Philippines
| |
Collapse
|
19
|
Olsson-Arvidsson L, Norberg A, Sjögren H, Johansson B. Frequent false-negative FIP1L1-PDGFRA FISH analyses of bone marrow samples from clonal eosinophilia at diagnosis. Br J Haematol 2019; 188:e76-e79. [PMID: 31854104 DOI: 10.1111/bjh.16340] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Linda Olsson-Arvidsson
- Department of Clinical Genetics and Pathology, Division of Laboratory Medicine, Skane University Hospital, Lund, Sweden
| | - Anna Norberg
- Medical Biosciences, Medical and Clinical Genetics, Umeå University, Umeå, Sweden
| | - Helene Sjögren
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bertil Johansson
- Department of Clinical Genetics and Pathology, Division of Laboratory Medicine, Skane University Hospital, Lund, Sweden.,Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
20
|
Shomali W, Gotlib J. World Health Organization-defined eosinophilic disorders: 2019 update on diagnosis, risk stratification, and management. Am J Hematol 2019; 94:1149-1167. [PMID: 31423623 DOI: 10.1002/ajh.25617] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of non-hematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia has generally been defined as a peripheral blood eosinophil count greater than 1.5 × 109 /L, and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of various tests. They include morphologic review of the blood and marrow, standard cytogenetics, fluorescence in situ-hybridization, flow immunophenotyping, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic hematolymphoid neoplasm. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2016 World Health Organization endorses a semi-molecular classification scheme of disease subtypes. This includes the major category "myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1 or with PCM1-JAK2", and the MPN subtype, "chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS). Lymphocyte-variant hypereosinophilia is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (eg, <1.5 × 109 /L) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant hypereosinophilia and HES. Hydroxyurea and interferon-alfa have demonstrated efficacy as initial treatment and in steroid-refractory cases of HES. In addition to hydroxyurea, second line cytotoxic chemotherapy agents, and hematopoietic stem cell transplantation have been used for aggressive forms of HES and CEL, with outcomes reported for limited numbers of patients. The use of antibodies against interleukin-5 (IL-5) (mepolizumab), the IL-5 receptor (benralizumab), as well as other targets on eosinophils remains an active area of investigation.
Collapse
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California
| | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, California
| |
Collapse
|
21
|
Xue Y, Cooper G, Cai C, Lu S, Hu B, Ma X, Lu X. Tumour-specific Causal Inference Discovers Distinct Disease Mechanisms Underlying Cancer Subtypes. Sci Rep 2019; 9:13225. [PMID: 31519988 PMCID: PMC6744493 DOI: 10.1038/s41598-019-48318-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/31/2019] [Indexed: 01/22/2023] Open
Abstract
Cancer is a disease mainly caused by somatic genome alterations (SGAs) that perturb cellular signalling systems. Furthermore, the combination of pathway aberrations in a tumour defines its disease mechanism, and distinct disease mechanisms underlie the inter-tumour heterogeneity in terms of disease progression and responses to therapies. Discovering common disease mechanisms shared by tumours would provide guidance for precision oncology but remains a challenge. Here, we present a novel computational framework for revealing distinct combinations of aberrant signalling pathways in tumours. Specifically, we applied the tumour-specific causal inference algorithm (TCI) to identify causal relationships between SGAs and differentially expressed genes (DEGs) within tumours from the Cancer Genome Atlas (TCGA) study. Based on these causal inferences, we adopted a network-based method to identify modules of DEGs, such that the member DEGs within a module tend to be co-regulated by a common pathway. Using the expression status of genes in a module as a surrogate measure of the activation status of the corresponding pathways, we divided breast cancers (BRCAs) into five subgroups and glioblastoma multiformes (GBMs) into six subgroups with distinct combinations of pathway aberrations. The patient groups exhibited significantly different survival patterns, indicating that our approach can identify clinically relevant disease subtypes.
Collapse
Affiliation(s)
- Yifan Xue
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Gregory Cooper
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Chunhui Cai
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States.,Paediatric Neurosurgery, UPMC Children's Hospital of Pittsburgh, Pittsburgh, 15213, United States.,Molecular and Cellular Cancer Biology Program, UPMC Hillman Cancer Centre, Pittsburgh, 15232, United States
| | - Xiaojun Ma
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, 15260, United States.
| |
Collapse
|
22
|
A complex and cryptic intrachromosomal rearrangement generating the FIP1L1_PDGFRA in adult acute myeloid leukemia. Cancer Genet 2019; 239:8-12. [PMID: 31450116 DOI: 10.1016/j.cancergen.2019.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/11/2019] [Accepted: 08/19/2019] [Indexed: 10/26/2022]
Abstract
Myeloid neoplasms with eosinophilia and abnormalities of the PDGFRA gene can benefit from therapy with tyrosine kinase inhibitors, therefore revealing the PDGFRA rearrangement is essential to ensure the best choice of treatment. The most common PDGFRA partner is the FIP1L1 gene, generating the oncoprotein FIP1L1/PDGFRA (F/P). In the majority of cases the F/P fusion gene originates from intrachromosomal rearrangement at band 4q12, and occasionally from chromosomal translocations. In both cases, the interstitial chromosomal deletion of a region involving the CHIC2 gene has been reported, which is cryptic by conventional karyotyping but detectable by Fluorescence In Situ Hybridization (FISH) analyses. Herein, we report an acute myeloid leukemia (AML) case presenting with eosinophilia; the F/P fusion gene originated from a new, cryptic and complex intrachromosomal rearrangement of 4q12. Classical FISH assay revealed abnormal hybridization signals, but the presence of the F/P chimaeric gene was demonstrated by molecular analysis. We performed molecular characterization of the chromosomal rearrangement and targeted Next-Generation Sequencing (NGS) analysis with a myeloid gene panel, revealing the presence of pathogenic genomic variants affecting the TET2 and ETV6 genes. These mutations were present as subclones at the disease onset and their clone size increased at relapse.
Collapse
|
23
|
Naymagon L, Marcellino B, Mascarenhas J. Eosinophilia in acute myeloid leukemia: Overlooked and underexamined. Blood Rev 2019; 36:23-31. [PMID: 30948162 DOI: 10.1016/j.blre.2019.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/05/2019] [Accepted: 03/28/2019] [Indexed: 02/04/2023]
Abstract
The presence of eosinophilia in acute myeloid leukemia (AML) suggests an underlying core binding factor (CBF) lesion, a platelet derived growth factor (PDGFR) translocation, or another rare translocation (such as ETV6-ABL1). Each of these cytogenetic entities carries unique diagnostic, prognostic, and therapeutic implications. CBF AML is most common and as such, its treatment is more clearly established, consisting of intensive induction chemotherapy followed by cytarabine based consolidation. Due in large part to its intrinsic chemo-sensitivity, CBF AML is associated with relatively high rates of remission and survival. PDGFR mediated AML is comparatively rare, and as such, diagnostic and treatment paradigms are not as well defined. Early identification of PDGFR translocations is essential, as they confer profound imatinib sensitivity which may, in many instances, spare the need for chemotherapy. Prompt recognition of such lesions requires a strong index of suspicion, and as such these diagnoses are often initially overlooked. Unfortunately, many cases of PDGFR associated AML, particularly those with other concurrent cytogenetic abnormalities, demonstrate treatment emergent imatinib resistance. Such patients continue to present a challenge, even with the advent of novel tyrosine kinase inhibitors (TKIs). Patients with rare translocations such as ETV6-ABL1 are not well described however seem to follow an aggressive clinical course, with limited response to imatinib, and poor outcomes. This review examines the significance of eosinophilia in the context of AML, with respect to its presentation, pathology, and cytogenetics, and with special attention to appropriate evaluation and treatment.
Collapse
Affiliation(s)
- Leonard Naymagon
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY 10029, USA.
| | - Bridget Marcellino
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY 10029, USA.
| | - John Mascarenhas
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1079, New York, NY 10029, USA.
| |
Collapse
|
24
|
Fang H, Ketterling RP, Hanson CA, Pardanani A, Kurtin PJ, Chen D, Greipp PT, Howard MT, King RL, Van Dyke DL, Reichard KK. A Test Utilization Approach to the Diagnostic Workup of Isolated Eosinophilia in Otherwise Morphologically Unremarkable Bone Marrow: A Single Institutional Experience. Am J Clin Pathol 2018; 150:421-431. [PMID: 30032299 DOI: 10.1093/ajcp/aqy064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Determine ancillary test utilization for the workup of isolated eosinophilia in otherwise morphologically unremarkable bone marrow (BM). METHODS We evaluated BM ancillary testing performed in cases with isolated eosinophilia and otherwise morphologically unremarkable BM. Cases with abnormal morphology (eg, dysplasia, basophilia) and/or findings suggestive of a disorder (eg, unexplained thromboses, lymphoma) are specifically excluded. RESULTS A total of 132 cases met inclusion criteria. Ten cases had an ancillary testing abnormality that warranted a more specific hematologic diagnosis: four cases of lymphocytic variant of hypereosinophilic syndrome, three cases of myeloid neoplasm with PDGFRA rearrangement, and one case each of myeloid neoplasm with PDGFRB rearrangement, chronic eosinophilic leukemia, and morphologically occult systemic mastocytosis. No cases revealed a cryptic PDGFRB or BCR/ABL1 rearrangement or JAK2 V617F mutation. CONCLUSIONS Findings from our institutional experience support initial testing in isolated eosinophilia with otherwise unremarkable BM to include PDGFRA rearrangement, tryptase/CD25 immunohistochemistry, cytogenetics, and T-cell flow cytometry/receptor gene rearrangement. This approach achieves diagnostic quality and test utilization efficiency in our clinical practice.
Collapse
Affiliation(s)
- Hong Fang
- Division of Hematopathology, Mayo Clinic, Rochester, MN
| | - Rhett P Ketterling
- Division of Laboratory Genetics and Genomics, Mayo Clinic, Rochester, MN
| | | | | | - Paul J Kurtin
- Division of Hematopathology, Mayo Clinic, Rochester, MN
| | - Dong Chen
- Division of Hematopathology, Mayo Clinic, Rochester, MN
| | - Patricia T Greipp
- Division of Laboratory Genetics and Genomics, Mayo Clinic, Rochester, MN
| | | | | | - Daniel L Van Dyke
- Division of Laboratory Genetics and Genomics, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
25
|
Abstract
Patients with PDGFRA-rearranged hematopoietic neoplasms typically present with chronic eosinophilic leukemia and rarely with acute myeloid leukemia or T-lymphoblastic lymphoma. However, mature T-cell lymphoma has not been previously associated with PDGFRA aberrations. We report a patient who presented with simultaneous T-lymphoblastic lymphoma, focal myeloid proliferation, and cutaneous cytotoxic T-cell lymphoma refractory to chemotherapy. The presence of myeloid and lymphoid lineages prompted genetic and molecular studies. A PDGFRA rearrangement was identified in all compartments: cutaneous, lymph node, and bone marrow. Treatment with imatinib resulted in an excellent response in cutaneous and systemic disease. We report the first case of a mature cutaneous T-cell lymphoma with PDGFRA rearrangement, expanding the spectrum of neoplasms associated with this genetic abnormality. Our case underscores the great importance of recognizing PDGFRA rearrangement in unusual cases of cutaneous T-cell lymphoma, systemic lymphoid, and myeloid neoplasms. These patients may respond well to tyrosine kinase inhibitors, despite resistance to conventional chemotherapy.
Collapse
|
26
|
Schneeweiss M, Peter B, Bibi S, Eisenwort G, Smiljkovic D, Blatt K, Jawhar M, Berger D, Stefanzl G, Herndlhofer S, Greiner G, Hoermann G, Hadzijusufovic E, Gleixner KV, Bettelheim P, Geissler K, Sperr WR, Reiter A, Arock M, Valent P. The KIT and PDGFRA switch-control inhibitor DCC-2618 blocks growth and survival of multiple neoplastic cell types in advanced mastocytosis. Haematologica 2018; 103:799-809. [PMID: 29439183 PMCID: PMC5927976 DOI: 10.3324/haematol.2017.179895] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
Systemic mastocytosis is a complex disease defined by abnormal growth and accumulation of neoplastic mast cells in various organs. Most patients exhibit a D816V-mutated variant of KIT, which confers resistance against imatinib. Clinical problems in systemic mastocytosis arise from mediator-related symptoms and/or organ destruction caused by malignant expansion of neoplastic mast cells and/or other myeloid cells in various organ systems. DCC-2618 is a spectrum-selective pan KIT and PDGFRA inhibitor which blocks KIT D816V and multiple other kinase targets relevant to systemic mastocytosis. We found that DCC-2618 inhibits the proliferation and survival of various human mast cell lines (HMC-1, ROSA, MCPV-1) as well as primary neoplastic mast cells obtained from patients with advanced systemic mastocytosis (IC50 <1 μM). Moreover, DCC-2618 decreased growth and survival of primary neoplastic eosinophils obtained from patients with systemic mastocytosis or eosinophilic leukemia, leukemic monocytes obtained from patients with chronic myelomonocytic leukemia with or without concomitant systemic mastocytosis, and blast cells obtained from patients with acute myeloid leukemia. Furthermore, DCC-2618 was found to suppress the proliferation of endothelial cells, suggesting additional drug effects on systemic mastocytosis-related angiogenesis. Finally, DCC-2618 was found to downregulate IgE-mediated histamine release from basophils and tryptase release from mast cells. Together, DCC-2618 inhibits growth, survival and activation of multiple cell types relevant to advanced systemic mastocytosis. Whether DCC-2618 is effective in vivo in patients with advanced systemic mastocytosis is currently under investigation in clinical trials.
Collapse
Affiliation(s)
- Mathias Schneeweiss
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Barbara Peter
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Siham Bibi
- Laboratoire de Biologie et Pharmacologie Appliquee, CNRS UMR 8113, Ecole Normale Superieure de Cachan, France
| | - Gregor Eisenwort
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | | | - Katharina Blatt
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Mohamad Jawhar
- Department of Hematology and Oncology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Daniela Berger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Susanne Herndlhofer
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Emir Hadzijusufovic
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria.,Department for Companion Animals and Horses, University Clinic for Small Animals, Internal Medicine Small Animals, University of Veterinary Medicine Vienna, Austria
| | - Karoline V Gleixner
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | | | - Klaus Geissler
- Fifth Medical Department, Hospital Hietzing, Vienna, Austria
| | - Wolfgang R Sperr
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| | - Andreas Reiter
- Department of Hematology and Oncology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Germany
| | - Michel Arock
- Laboratoire de Biologie et Pharmacologie Appliquee, CNRS UMR 8113, Ecole Normale Superieure de Cachan, France
| | - Peter Valent
- Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria .,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Austria
| |
Collapse
|
27
|
Hilal T, Fauble V, Ketterling RP, Kelemen K. Myeloid neoplasm with eosinophilia associated with isolated extramedullary FIP1L1 / PDGFRA rearrangement. Cancer Genet 2018; 220:13-18. [DOI: 10.1016/j.cancergen.2017.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 10/16/2017] [Indexed: 11/17/2022]
|
28
|
Myeloid and Lymphoid Neoplasms with Eosinophilia and Abnormalities of PDGFRA, PDGFRB, FGFR1, or t(8;9)(p22;p24.1);PCM1-JAK2. MOLECULAR PATHOLOGY LIBRARY 2018. [DOI: 10.1007/978-3-319-62146-3_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
29
|
Identification of autofluorescent cells in human angioimmunoblastic T-cell lymphoma. Histochem Cell Biol 2017; 149:169-177. [PMID: 29197996 DOI: 10.1007/s00418-017-1624-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 02/08/2023]
Abstract
Endogenous cell autofluorescence is a common nuisance that complicates the use of fluorescence microscopy. When using fluorescence-labeled antibodies for specific cell labeling in tissue sections of human angioimmunoblastic T-cell lymphoma (AITL), we encountered with a problematic autofluorescence of multiple cells. These cells emitted fluorescence signals in the green, red and deep-red spectral range. Characterization of these autofluorescent cells solely on the basis of their autofluorescence failed. To identify these enigmatic cells residing the lymphoma tissue, we combined two imaging techniques-fluorescence and brightfield microscopy. Combined fluorescence/brightfield imaging of cells immunolabeled with a panel of CD antibodies raised against diverse cellular components allowed us to identify the autofluorescent cells in the AITL as eosinophils. These cells tended to accumulate in the vicinity of capillaries and arterioles apparently mediating the process of angiogenesis resembling other angiogenesis-associated diseases.
Collapse
|
30
|
Gotlib J. World Health Organization-defined eosinophilic disorders: 2017 update on diagnosis, risk stratification, and management. Am J Hematol 2017; 92:1243-1259. [PMID: 29044676 DOI: 10.1002/ajh.24880] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 12/20/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of nonhematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia has generally been defined as a peripheral blood eosinophil count greater than 1500/mm3 and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of morphologic review of the blood and marrow, standard cytogenetics, fluorescent in situ-hybridization, flow immunocytometry, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic myeloid or lymphoproliferative disorder. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2016 World Health Organization endorses a semi-molecular classification scheme of disease subtypes which includes the major category "myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1 or with PCM1-JAK2," and the "MPN subtype, chronic eosinophilic leukemia, not otherwise specified" (CEL, NOS). Lymphocyte-variant hypereosinophilia is an aberrant T-cell clone-driven reactive eosinophila, and idiopathic hypereosinophilic syndrome (HES) is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g., < 1500/mm3 ) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant hypereosinophilia and HES. Hydroxyurea and interferon-alpha have demonstrated efficacy as initial treatment and steroid-refractory cases of HES. In addition to hydroxyurea, second line cytotoxic chemotherapy agents and hematopoietic cell transplant have been used for aggressive forms of HES and CEL with outcomes reported for limited numbers of patients. The use of antibodies against interleukin-5 (IL-5) (mepolizumab), the IL-5 receptor (benralizumab), and CD52 (alemtuzumab), as well as other targets on eosinophils remains an active area of investigation.
Collapse
Affiliation(s)
- Jason Gotlib
- Stanford Cancer Institute, Stanford, California 94305-5821
| |
Collapse
|
31
|
Scherber RM, Borate U. How we diagnose and treat systemic mastocytosis in adults. Br J Haematol 2017; 180:11-23. [DOI: 10.1111/bjh.14967] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Robyn M. Scherber
- Department of Hematology and Oncology; Oregon Health and Science University; Portland OR USA
- Department of Hematology and Oncology; Mayo Clinic; Scottsdale AZ USA
| | - Uma Borate
- Department of Hematology and Oncology; Oregon Health and Science University; Portland OR USA
| |
Collapse
|
32
|
Gotlib J. Tyrosine Kinase Inhibitors in the Treatment of Eosinophilic Neoplasms and Systemic Mastocytosis. Hematol Oncol Clin North Am 2017; 31:643-661. [PMID: 28673393 DOI: 10.1016/j.hoc.2017.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The World Health Organization's semimolecular classification of eosinophilias emphasizes neoplasms driven by fusion tyrosine kinases. More than 80% of patients with systemic mastocytosis carry the KIT D816V mutation, the primary driver of disease pathogenesis. Genetic annotation of these diseases is critical and affords opportunities for targeted therapy. This article discusses our understanding of the mutated tyrosine kinome of eosinophilic neoplasms and systemic mast cell disease, and the successes and limitations of available therapies. Use of tyrosine kinase inhibitors as a bridge to hematopoietic stem cell transplantation, and development of more selective and potent tyrosine kinase inhibitors is also highlighted.
Collapse
Affiliation(s)
- Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, 875 Blake Wilbur Drive, Room 2324, Stanford, CA 94305-5821, USA.
| |
Collapse
|
33
|
Ishii Y, Hamashima T, Yamamoto S, Sasahara M. Pathogenetic significance and possibility as a therapeutic target of platelet derived growth factor. Pathol Int 2017; 67:235-246. [DOI: 10.1111/pin.12530] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/27/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Yoko Ishii
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| | - Takeru Hamashima
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| | - Seiji Yamamoto
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| | - Masakiyo Sasahara
- Department of Pathology; Graduate School of Medicine and Pharmaceutical Sciences; University of Toyama; Toyama 930-0194 Japan
| |
Collapse
|
34
|
Abstract
Abstract
Molecular diagnostics has generated substantial dividends in dissecting the genetic basis of myeloid neoplasms with eosinophilia. The family of diseases generated by dysregulated fusion tyrosine kinase (TK) genes is recognized by the World Health Organization (WHO) category, “Myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1, or with PCM1-JAK2.” In addition to myeloproliferative neoplasms (MPN), these patients can present with myelodysplastic syndrome/MPN, as well as de novo or secondary mixed-phenotype leukemias or lymphomas. Eosinophilia is a common, but not invariable, feature of these diseases. The natural history of PDGFRA- and PDGFRB-rearranged neoplasms has been dramatically altered by imatinib. In contrast, patients with FGFR1 and JAK2 fusion TK genes exhibit a more aggressive course and variable sensitivity to current TK inhibitors, and in most cases, long-term disease-free survival may only be achievable with allogeneic hematopoietic stem cell transplantation. Similar poor prognosis outcomes may be observed with rearrangements of FLT3 or ABL1 (eg, both of which commonly partner with ETV6), and further investigation is needed to validate their inclusion in the current WHO-defined group of eosinophilia-associated TK fusion-driven neoplasms. The diagnosis chronic eosinophilic leukemia, not otherwise specified (CEL, NOS) is assigned to patients with MPN with eosinophilia and nonspecific cytogenetic/molecular abnormalities and/or increased myeloblasts. Myeloid mutation panels have identified somatic variants in patients with a provisional diagnosis of hypereosinophilia of undetermined significance, reclassifying some of these cases as eosinophilia-associated neoplasms. Looking forward, one of the many challenges will be how to use the results of molecular profiling to guide prognosis and selection of actionable therapeutic targets.
Collapse
|
35
|
Abstract
Eosinophils can regulate local and systemic inflammation, and their presence in higher numbers appears to play an important role in the pathology of various atopic and inflammatory diseases. Eosinophil maturation, recruitment, and survival depend on several cytokine regulators, including interleukin (IL)-5, IL-4, and IL-13 as well as growth factors such as GM-CSF. Over the last decade, the approach to treating eosinophilic diseases has changed greatly. A number of biologic modulators have been developed to target eosinophilic inflammatory pathways, and their usage has resulted in variable clinical improvement in the treatment of eosinophilic-associated conditions. Novel targeted therapies that are safe and effective for treating these disorders are being investigated. This review summarizes the clinical use of biologic agents that have been studied in clinical trials or approved for treating eosinophilic diseases.
Collapse
Affiliation(s)
- Panida Sriaroon
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of South Florida, 140 7th ave S, CRI 4008, St. Petersburg, FL, 33701, USA.
| | - Mark Ballow
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of South Florida, 140 7th ave S, CRI 4008, St. Petersburg, FL, 33701, USA
| |
Collapse
|
36
|
Nelson KN, Peiris MN, Meyer AN, Siari A, Donoghue DJ. Receptor Tyrosine Kinases: Translocation Partners in Hematopoietic Disorders. Trends Mol Med 2016; 23:59-79. [PMID: 27988109 DOI: 10.1016/j.molmed.2016.11.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/11/2016] [Accepted: 11/13/2016] [Indexed: 02/07/2023]
Abstract
Receptor tyrosine kinases (RTKs) activate various signaling pathways and regulate cellular proliferation, survival, migration, and angiogenesis. Malignant neoplasms often circumvent or subjugate these pathways by promoting RTK overactivation through mutation or chromosomal translocation. RTK translocations create a fusion protein containing a dimerizing partner fused to an RTK kinase domain, resulting in constitutive kinase domain activation, altered RTK cellular localization, upregulation of downstream signaling, and novel pathway activation. While RTK translocations in hematological malignancies are relatively rare, clinical evidence suggests that patients with these genetic abnormalities benefit from RTK-targeted inhibitors. Here, we present a timely review of an exciting field by examining RTK chromosomal translocations in hematological cancers, such as Anaplastic Lymphoma Kinase (ALK), Fibroblast Growth Factor Receptor (FGFR), Platelet-Derived Growth Factor Receptor (PDGFR), REarranged during Transfection (RET), Colony Stimulating Factor 1 Receptor (CSF1R), and Neurotrophic Tyrosine Kinase Receptor Type 3 (NTRK3) fusions, and discuss current therapeutic options.
Collapse
Affiliation(s)
- Katelyn N Nelson
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Malalage N Peiris
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - April N Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Asma Siari
- Université Joseph Fourier Grenoble, Grenoble, France
| | - Daniel J Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA; Moores UCSD Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
37
|
Pardanani A. Systemic mastocytosis in adults: 2017 update on diagnosis, risk stratification and management. Am J Hematol 2016; 91:1146-1159. [PMID: 27762455 DOI: 10.1002/ajh.24553] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/12/2016] [Indexed: 12/17/2022]
Abstract
Disease overview:Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extra-cutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of morphologically abnormal MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC expression of CD25 and/or CD2, and presence of KITD816V. Risk stratification: The 2008 World Health Organization (WHO) classification of SM has been shown to be prognostically relevant. Classification of SM patients into indolent (SM), aggressive SM (ASM), SM associated with a clonal non-MC lineage disease (SM-AHNMD) and mast cell leukemia (MCL) subgroups is a useful first step in establishing prognosis. MANAGEMENT SM treatment is generally palliative. ISM patients have a normal life expectancy and receive symptom-directed therapy; infrequently, cytoreductive therapy may be indicated for refractory symptoms. ASM patients have disease-related organ dysfunction; interferon-α (±corticosteroids) can control dermatological, hematological, gastrointestinal, skeletal and mediator-release symptoms, but is hampered by poor tolerability. Similarly, cladribine has broad therapeutic activity, with particular utility when rapid MC debulking is indicated; the main toxicity is myelosuppression. Imatinib has a therapeutic role in the presence of an imatinib-sensitive KIT mutation or in KITD816-unmutated patients. Treatment of SM-AHNMD is governed primarily by the non-MC neoplasm; hydroxyurea has modest utility in this setting; there is a role for allogeneic stem cell transplantation in select cases. Investigational drugs: Recent data confirms midostaurin's significant anti-MC activity in patients with advanced SM. Am. J. Hematol. 91:1147-1159, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
38
|
Alvarez-Twose I, Matito A, Morgado JM, Sánchez-Muñoz L, Jara-Acevedo M, García-Montero A, Mayado A, Caldas C, Teodósio C, Muñoz-González JI, Mollejo M, Escribano L, Orfao A. Imatinib in systemic mastocytosis: a phase IV clinical trial in patients lacking exon 17 KIT mutations and review of the literature. Oncotarget 2016; 8:68950-68963. [PMID: 28978170 PMCID: PMC5620310 DOI: 10.18632/oncotarget.10711] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/29/2016] [Indexed: 12/15/2022] Open
Abstract
Resistance to imatinib has been recurrently reported in systemic mastocytosis (SM) carrying exon 17 KIT mutations. We evaluated the efficacy and safety of imatinib therapy in 10 adult SM patients lacking exon 17 KIT mutations, 9 of which fulfilled criteria for well-differentiated SM (WDSM). The World Health Organization 2008 disease categories among WDSM patients were mast cell (MC) leukemia (n = 3), indolent SM (n = 3) and cutaneous mastocytosis (n = 3); the remainder case had SM associated with a clonal haematological non-MC disease. Patients were given imatinib for 12 months -400 or 300 mg daily depending on the presence vs. absence of > 30% bone marrow (BM) MCs and/or signs of advanced disease-. Absence of exon 17 KIT mutations was confirmed in highly-purified BM MCs by peptide nucleic acid-mediated PCR, while mutations involving other exons were investigated by direct sequencing of purified BM MC DNA. Complete response (CR) was defined as resolution of BM MC infiltration, skin lesions, organomegalies and MC-mediator release-associated symptoms, plus normalization of serum tryptase. Criteria for partial response (PR) included ≥ 50% reduction in BM MC infiltration and improvement of skin lesions and/or organomegalies. Treatment was well-tolerated with an overall response rate of 50%, including early and sustained CR in four patients, three of whom had extracellular mutations of KIT, and PR in one case. This later patient and all non-responders (n = 5) showed wild-type KIT. These results together with previous data from the literature support the relevance of the KIT mutational status in selecting SM patients who are candidates for imatinib therapy.
Collapse
Affiliation(s)
- Iván Alvarez-Twose
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Hospital Virgen del Valle, Toledo, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Almudena Matito
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Hospital Virgen del Valle, Toledo, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - José Mário Morgado
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Hospital Virgen del Valle, Toledo, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Laura Sánchez-Muñoz
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast), Hospital Virgen del Valle, Toledo, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - María Jara-Acevedo
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Andrés García-Montero
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Andrea Mayado
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Carolina Caldas
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Cristina Teodósio
- Department of Immunology, Erasmus Medical Center, University of Rotterdam, Rotterdam, The Netherlands
| | - Javier Ignacio Muñoz-González
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Manuela Mollejo
- Department of Pathology, Hospital Virgen de la Salud, Toledo, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Luis Escribano
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| | - Alberto Orfao
- Centro de Investigación del Cáncer/IBMCC (USAL/CSIC) and IBSAL, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.,Spanish Network on Mastocytosis (REMA), Toledo and Salamanca, Spain
| |
Collapse
|
39
|
Blood's 70th anniversary: musings of a Blood editor, 2003-2007. Blood 2016; 127:2649-51. [PMID: 27257172 DOI: 10.1182/blood-2015-11-635755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 12/03/2015] [Indexed: 11/20/2022] Open
|
40
|
Abstract
Eosinophilia in the peripheral blood can be the manifestation various medical conditions, including benign or malignant disorders. There are 3 main types of eosinophilia-associated myeloid neoplasms (MN-eos): myeloid and lymphoid neoplasms, chronic eosinophilic leukemia not otherwise specified, and idiopathic hypereosinophilic syndrome (HES). Imatinib mesylate has revolutionized the treatment of molecularly defined MN-eos, and novel agents have been successfully used to treat HES. The discovery of new, recurrent molecular alterations in patients with MN-eos may improve their diagnosis and therapy. This review focuses on the hematologist's approach to a patient with eosinophilia and treatment options for those with eosinophilic myeloid neoplasms.
Collapse
Affiliation(s)
- Lorenzo Falchi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Gotlib J. World Health Organization-defined eosinophilic disorders: 2015 update on diagnosis, risk stratification, and management. Am J Hematol 2015; 90:1077-89. [PMID: 26486351 DOI: 10.1002/ajh.24196] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 12/19/2022]
Abstract
DISEASE OVERVIEW The eosinophilias encompass a broad range of non-hematologic (secondary or reactive) and hematologic (primary, clonal) disorders with potential for end-organ damage. DIAGNOSIS Hypereosinophilia has generally been defined as a peripheral blood eosinophil count greater than 1,500/mm(3) and may be associated with tissue damage. After exclusion of secondary causes of eosinophilia, diagnostic evaluation of primary eosinophilias relies on a combination of morphologic review of the blood and marrow, standard cytogenetics, fluorescent in situ-hybridization, flow immunocytometry, and T-cell clonality assessment to detect histopathologic or clonal evidence for an acute or chronic myeloid or lymphoproliferative disorder. RISK STRATIFICATION Disease prognosis relies on identifying the subtype of eosinophilia. After evaluation of secondary causes of eosinophilia, the 2008 World Health Organization establishes a semi-molecular classification scheme of disease subtypes including 'myeloid and lymphoid neoplasms with eosinophilia and abnormalities of PDGFRA, PDGFRB, or FGFR1', chronic eosinophilic leukemia, not otherwise specified, (CEL, NOS), lymphocyte-variant hypereosinophilia, and idiopathic hypereosinophilic syndrome (HES), which is a diagnosis of exclusion. RISK-ADAPTED THERAPY The goal of the therapy is to mitigate eosinophil-mediated organ damage. For patients with milder forms of eosinophilia (e.g. < 1,500/mm(3) ) without symptoms or signs of organ involvement, a watch and wait approach with close-follow-up may be undertaken. Identification of rearranged PDGFRA or PDGFRB is critical because of the exquisite responsiveness of these diseases to imatinib. Corticosteroids are first-line therapy for patients with lymphocyte-variant hypereosinophilia and HES. Hydroxyurea and interferon-alpha have demonstrated efficacy as initial treatment and steroid-refractory cases of HES. In addition to hydroxyurea, second line cytotoxic chemotherapy agents and hematopoietic cell transplant have been used for aggressive forms of HES and CEL with outcomes reported for limited numbers of patients. Although clinical trials have been performed with anti IL-5 (mepolizumab) and anti-CD52 (alemtuzumab) antibodies, their therapeutic role in primary eosinophilic diseases and HES has yet to be established.
Collapse
|
42
|
Bonkobara M. Dysregulation of tyrosine kinases and use of imatinib in small animal practice. Vet J 2015; 205:180-8. [DOI: 10.1016/j.tvjl.2014.12.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 12/01/2014] [Accepted: 12/16/2014] [Indexed: 12/18/2022]
|
43
|
Kim JA, Im KO, Park SN, Kwon JS, Kim SY, Oh K, Lee DS, Kim MK, Kim SW, Jang M, Lee G, Oh YM, Lee SD, Lee DS. Cytogenetic heterogeneity and their serial dynamic changes during acquisition of cytogenetic aberrations in cultured mesenchymal stem cells. Mutat Res 2015; 777:60-8. [PMID: 25974687 DOI: 10.1016/j.mrfmmm.2015.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 04/02/2015] [Accepted: 04/06/2015] [Indexed: 12/25/2022]
Abstract
To minimize the risk of tumorigenesis in mesenchymal stem cells (MSCs), G-banding analysis is widely used to detect chromosomal aberrations in MSCs. However, a critical limitation of G-banding is that it only reflects the status of metaphase cells, which can represent as few as 0.01% of tested cells. During routine cytogenetic testing in MSCs, we often detect chromosomal aberrations in minor cell populations. Therefore, we aimed to investigate whether such a minority of cells can expand over time or if they ultimately disappear during MSC passaging. We passaged MSCs serially while monitoring quantitative changes for each aberrant clone among heterogeneous MSCs. To investigate the cytogenetic status of interphase cells, which represent the main population, we also performed interphase FISH analysis, in combination with G-banding and telomere length determination. In human adipose tissue-derived MSCs, 4 types of chromosomal aberrations were found during culturing, and in umbilical cord MSCs, 2 types of chromosomal aberrations were observed. Sequential dynamic changes among heterogeneous aberrant clones during passaging were similar to the dynamic changes observed in cancer stem cells during disease progression. Throughout all passages, the quantitative G-banding results were inconsistent with those of the interphase FISH analysis. Interphase FISH revealed hidden aberrations in stem cell populations with normal karyotypes by G-banding analysis. We found that telomere length gradually decreased during passaging until the point at which cytogenetic aberrations appeared. The present study demonstrates that rare aberrant clones at earlier passages can become predominant clones during later passages. Considering the risk of tumorigenesis due to aberrant MSCs, we believe that our results will help to establish proper safety guidelines for MSC use. In particular, we believe it is critical to test for chromosomal aberrations using both G-banding and FISH to ensure the safety of human stem cells that are manufactured in vitro for clinical applications.
Collapse
Affiliation(s)
- Jung-Ah Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyong Ok Im
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Si Nae Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ji Seok Kwon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seon Young Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keunhee Oh
- Laboratory of Immunology and Cancer Biology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Transplantation Research Institute, Seoul National University College of Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong-Sup Lee
- Laboratory of Immunology and Cancer Biology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea; Transplantation Research Institute, Seoul National University College of Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Min Kyung Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Jang
- Lab of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Gene Lee
- Lab of Molecular Genetics, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asthma Center and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Do Lee
- Department of Pulmonary and Critical Care Medicine, Asthma Center and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong Soon Lee
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Shin SY, Jung CW, Choi DC, Lee BJ, Kim HJ, Kim SH. Chronic eosinophilic leukemia with a FIP1L1-PDGFRA rearrangement: Two case reports and a review of Korean cases. Blood Res 2015; 50:58-61. [PMID: 25830134 PMCID: PMC4377343 DOI: 10.5045/br.2015.50.1.58] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 08/05/2014] [Accepted: 02/12/2015] [Indexed: 11/22/2022] Open
Affiliation(s)
- Sang-Yong Shin
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chul-Won Jung
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Chull Choi
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung-Jae Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sun-Hee Kim
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
45
|
Chisholm KM, Merker JD, Gotlib JR, Gitana G, Lefterova M, Zehnder JL, George TI, Arber DA, Ohgami RS. Mast cells in systemic mastocytosis have distinctly brighter CD45 expression by flow cytometry. Am J Clin Pathol 2015; 143:527-34. [PMID: 25780004 DOI: 10.1309/ajcpz3j4geeyirra] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES We sought to determine the significance of bright CD45 expression on mast cells in cases of systemic mastocytosis vs mast cells in bone marrows uninvolved by systemic mastocytosis and compare this CD45 expression with CD25 and CD2 expression on mast cells. METHODS Multiparameter flow cytometry was performed on 31 cases of systemic mastocytosis and 70 bone marrow cases that were not involved by systemic mastocytosis. Bright expression of CD45 was defined as more than 20% of CD117+ mast cells showing brighter CD45 expression than the average expression level of lymphocytes. RESULTS Mast cells with bright CD45 expression were seen in 26 systemic mastocytosis cases and three bone marrows uninvolved by systemic mastocytosis (sensitivity, 84%; specificity, 96%). CD25 alone had a greater sensitivity (100%) but lower specificity (93%) compared with bright CD45 for identifying abnormal mast cells, while CD2 alone had lower sensitivity but higher specificity. To reach a specificity of 100%, CD25 together with bright CD45 on mast cells was the optimal combination to detect cases of systemic mastocytosis. CONCLUSIONS A combination of bright CD45 and CD25 appears to specifically identify abnormal mast cells in cases of systemic mastocytosis. Further studies will be necessary to confirm these results.
Collapse
Affiliation(s)
- Karen M. Chisholm
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Jason D. Merker
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Jason R. Gotlib
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Gary Gitana
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Martina Lefterova
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - James L. Zehnder
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Tracy I. George
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque
| | - Daniel A. Arber
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Robert S. Ohgami
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
46
|
Pardanani A. Systemic mastocytosis in adults: 2015 update on diagnosis, risk stratification, and management. Am J Hematol 2015; 90:250-62. [PMID: 25688753 DOI: 10.1002/ajh.23931] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022]
Abstract
DISEASE OVERVIEW Systemic mastocytosis (SM) results from a clonal proliferation of abnormal mast cells (MC) in one or more extracutaneous organs. DIAGNOSIS The major criterion is presence of multifocal clusters of morphologically abnormal MC in the bone marrow. Minor diagnostic criteria include elevated serum tryptase level, abnormal MC expression of CD25 and/or CD2, and presence of KITD816V. RISK STRATIFICATION The 2008 World Health Organization classification of SM has been shown to be prognostically relevant. Classification of SM patients into indolent SM (ISM), aggressive SM (ASM), SM associated with a clonal non-MC lineage disease (SM-AHNMD), and mast cell leukemia (MCL) subgroups is a useful first step in establishing prognosis. MANAGEMENT SM treatment is generally palliative. ISM patients have a normal life expectancy and receive symptom-directed therapy; infrequently, cytoreductive therapy may be indicated for refractory symptoms. ASM patients have disease-related organ dysfunction; interferon-α (+/-corticosteroids) can control dermatological, hematological, gastrointestinal, skeletal, and mediator-release symptoms, but is hampered by poor tolerability. Similarly, cladribine has broad therapeutic activity, with particular utility when rapid MC debulking is indicated; the main toxicity is myelosuppression. Imatinib has a therapeutic role in the presence of an imatinib-sensitive KIT mutation or in KITD816-unmutated patients. Treatment of SM-AHNMD is governed primarily by the non-MC neoplasm; hydroxyurea has modest utility in this setting; there is a role for allogeneic stem cell transplantation in select cases. Investigational Drugs: Recent data confirms midostaurin's significant anti-MC activity in patients with advanced SM.
Collapse
Affiliation(s)
- Animesh Pardanani
- Division of Hematology; Mayo Clinic; Rochester Minnesota
- Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
47
|
Chalmers ZR, Ali SM, Ohgami RS, Campregher PV, Frampton GM, Yelensky R, Elvin JA, Palma NA, Erlich R, Vergilio JA, Chmielecki J, Ross JS, Stephens PJ, Hermann R, Miller VA, Miles CR. Comprehensive genomic profiling identifies a novel TNKS2-PDGFRA fusion that defines a myeloid neoplasm with eosinophilia that responded dramatically to imatinib therapy. Blood Cancer J 2015; 5:e278. [PMID: 25658984 PMCID: PMC4349257 DOI: 10.1038/bcj.2014.95] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
| | - S M Ali
- Foundation Medicine Inc., Cambridge, MA, USA
| | - R S Ohgami
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - P V Campregher
- 1] Foundation Medicine Inc., Cambridge, MA, USA [2] Clinical Laboratory, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | - R Yelensky
- Foundation Medicine Inc., Cambridge, MA, USA
| | - J A Elvin
- Foundation Medicine Inc., Cambridge, MA, USA
| | - N A Palma
- Foundation Medicine Inc., Cambridge, MA, USA
| | - R Erlich
- Foundation Medicine Inc., Cambridge, MA, USA
| | - J-A Vergilio
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - J S Ross
- 1] Foundation Medicine Inc., Cambridge, MA, USA [2] Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY, USA
| | | | - R Hermann
- Northwest Georgia Oncology Centers, Jasper Cancer Center, Jasper, GA, USA
| | - V A Miller
- Foundation Medicine Inc., Cambridge, MA, USA
| | - C R Miles
- Northwest Georgia Oncology Centers, Jasper Cancer Center, Jasper, GA, USA
| |
Collapse
|
48
|
Nemchenko IS, Turkina AG, Chelysheva EY, Galstyan GM, Kovrigina AM, Khuazheva NK, Savchenko VG. [FIP1L1-PDGFRА-positive myeloproliferative disease with eosinophilia: A rare case with multiple organ dysfunction and a response to tyrosine kinase inhibitor therapy]. TERAPEVT ARKH 2015; 87:89-95. [PMID: 26978425 DOI: 10.17116/terarkh2015871289-95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The described case of FIP1L1-PDGFRА-positive myeloproliferative disease is characterized by an atypical aggressive course to develop severe specific complications as injuries to the brain, heart, lung, and intestine. Pathogenetic therapy with imatinib could stabilize a patient's state, but failed to produce a complete hematological response. Switching from imatinib to dasatinib could produce sustained clinical, hematological, and molecular remissions.
Collapse
Affiliation(s)
- I S Nemchenko
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A G Turkina
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - E Yu Chelysheva
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - G M Galstyan
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - A M Kovrigina
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| | - N K Khuazheva
- S.P. Botkin City Clinical Hospital, Moscow Healthcare Department, Moscow, Russia
| | - V G Savchenko
- Hematology Research Center, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
49
|
Pautier P, Locher C, Robert C, Deroussent A, Flament C, Le Cesne A, Rey A, Bahleda R, Ribrag V, Soria JC, Vassal G, Eggermont A, Zitvogel L, Chaput N, Paci A. Phase I clinical trial combining imatinib mesylate and IL-2 in refractory cancer patients: IL-2 interferes with the pharmacokinetics of imatinib mesylate. Oncoimmunology 2014; 2:e23079. [PMID: 23525192 PMCID: PMC3601177 DOI: 10.4161/onci.23079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Imatinib mesylate (IM) is a small molecule inhibitor of protein tyrosine kinases. In addition to its direct effect on malignant cells, it has been suggested IM may activate of natural killer (NK) cells, hence exerting immunomodulatory functions. In preclinical settings, improved antitumor responses have been observed when IM and interleukin-2 (IL-2), a cytokine that enhances NK cells functions, were combined. The goals of this study were to determine the maximum tolerated dose (MTD) of IL-2 combined with IM at a constant dose of 400 mg, the pharmacokinetics of IM and IL-2, as well as toxicity and clinical efficacy of this immunotherapeutic regimen in patients affected by advanced tumors. The treatment consisted in 50 mg/day cyclophosphamide from 21 d before the initiation of IM throughout the first IM cycle (from D-21 to D14), 400 mg/day IM for 14 d (D1 to D14) combined with escalating doses of IL-2 (3, 6, 9 and 12 MIU/day) from days 10 to 14. This treatment was administered at three week intervals to 17 patients. Common side effects of the combination were mild to moderate, including fever, chills, fatigue, nausea and hepatic enzyme elevation. IL-2 dose level II, 6 MIU/day, was determined as the MTD with the following dose-limiting toxicities: systemic capillary leak syndrome, fatigue and anorexia. Pharmacokinetic studies revealed that the area under the curve and the maximum concentration of IM and its main metabolite CGP74588 increased significantly when IM was concomitantly administered with IL-2. In contrast, IM did not modulate IL-2 pharmacokinetics. No objective responses were observed. The best response obtained was stable disease in 8/17 (median duration: 12 weeks). Finally, IL-2 augmented the impregnation of IM and its metabolite. The combination of IM (400 mg/day) and IL-2 (6 MIU/day) in tumors that express IM targets warrants further investigation.
Collapse
Affiliation(s)
- Patricia Pautier
- Département de Médecine; Institut de Cancérologie Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Aypar U, Knudson RA, Pearce KE, Wiktor AE, Ketterling RP. Development of an NPM1/MLF1 D-FISH Probe Set for the Detection of t(3;5)(q25;q35) Identified in Patients with Acute Myeloid Leukemia. J Mol Diagn 2014; 16:527-532. [DOI: 10.1016/j.jmoldx.2014.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/25/2014] [Accepted: 05/16/2014] [Indexed: 11/27/2022] Open
|