1
|
Cilloni D, Maffeo B, Savi A, Danzero AC, Bonuomo V, Fava C. Detection of KIT Mutations in Systemic Mastocytosis: How, When, and Why. Int J Mol Sci 2024; 25:10885. [PMID: 39456668 PMCID: PMC11507058 DOI: 10.3390/ijms252010885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/28/2024] Open
Abstract
More than 90% of patients affected by mastocytosis are characterized by a somatic point mutation of KIT, which induces ligand-independent activation of the receptor and downstream signal triggering, ultimately leading to mast cell accumulation and survival. The most frequent mutation is KIT p.D816V, but other rarer mutations can also be found. These mutations often have a very low variant allele frequency (VAF), well below the sensitivity of common next-generation sequencing (NGS) methods used in routine diagnostic panels. Highly sensitive methods are developing for detecting mutations. This review summarizes the current indications on the recommended methods and on how to manage and interpret molecular data for the diagnosis and follow-up of patients with mastocytosis.
Collapse
Affiliation(s)
- Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Mauriziano Hospital, 10128 Turin, Italy; (B.M.); (A.S.); (A.C.D.); (V.B.); (C.F.)
| | | | | | | | | | | |
Collapse
|
2
|
Murray HC, Miller K, Dun MD, Verrills NM. Pharmaco-phosphoproteomic analysis of cancer-associated KIT mutations D816V and V560G. Proteomics 2024; 24:e2300309. [PMID: 38334196 DOI: 10.1002/pmic.202300309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/24/2023] [Accepted: 01/17/2024] [Indexed: 02/10/2024]
Abstract
The CD117 mast/stem cell growth factor receptor tyrosine kinase (KIT) is critical for haematopoiesis, melanogenesis and stem cell maintenance. KIT is commonly activated by mutation in cancers including acute myeloid leukaemia, melanoma and gastrointestinal stromal tumours (GISTs). The kinase and the juxtamembrane domains of KIT are mutation hotspots; with the kinase domain mutation D816V common in leukaemia and the juxtamembrane domain mutation V560G common in GISTs. Given the importance of mutant KIT signalling in cancer, we have conducted a proteomic and phosphoproteomic analysis of myeloid progenitor cells expressing D816V- and V560G-KIT mutants, using an FDCP1 isogenic cell line model. Proteomic analysis revealed increased abundance of proteases and growth signalling proteins in KIT-mutant cells compared to empty vector (EV) controls. Pathway analysis identified increased oxidative phosphorylation in D816V- and V560G-mutant KIT cells, which was targetable using the inhibitor IACS010759. Dysregulation of RNA metabolism and cytoskeleton/adhesion pathways was identified in both the proteome and phosphoproteome of KIT-mutant cells. Phosphoproteome analysis further revealed active kinases such as EGFR, ERK and PKC, which were targetable using pharmacological inhibitors. This study provides a pharmaco-phosphoproteomic profile of D816V- and V560G-mutant KIT cells, which reveals novel therapeutic strategies that may be applicable to a range of cancers.
Collapse
Affiliation(s)
- Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Kasey Miller
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, and Precision Medicine Program, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
3
|
Sevilla A, Grichnik J. Therapeutic modulation of KIT ligand in melanocytic disorders with implications for mast cell diseases. Exp Dermatol 2024; 33:e15091. [PMID: 38711220 DOI: 10.1111/exd.15091] [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: 11/20/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/08/2024]
Abstract
KIT ligand and its associated receptor KIT serve as a master regulatory system for both melanocytes and mast cells controlling survival, migration, proliferation and activation. Blockade of this pathway results in cell depletion, while overactivation leads to mastocytosis or melanoma. Expression defects are associated with pigmentary and mast cell disorders. KIT ligand regulation is complex but efficient targeting of this system would be of significant benefit to those suffering from melanocytic or mast cell disorders. Herein, we review the known associations of this pathway with cutaneous diseases and the regulators of this system both in skin and in the more well-studied germ cell system. Exogenous agents modulating this pathway will also be presented. Ultimately, we will review potential therapeutic opportunities to help our patients with melanocytic and mast cell disease processes potentially including vitiligo, hair greying, melasma, urticaria, mastocytosis and melanoma.
Collapse
Affiliation(s)
- Alec Sevilla
- Department of Dermatology, New York Medical College, New York, New York, USA
- Department of Internal Medicine, Lakeland Regional Health, Lakeland, Florida, USA
| | - James Grichnik
- Department of Dermatology and Cutaneous Surgery, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
4
|
Ahmed N, Preisinger C, Wilhelm T, Huber M. TurboID-Based IRE1 Interactome Reveals Participants of the Endoplasmic Reticulum-Associated Protein Degradation Machinery in the Human Mast Cell Leukemia Cell Line HMC-1.2. Cells 2024; 13:747. [PMID: 38727283 PMCID: PMC11082977 DOI: 10.3390/cells13090747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
The unfolded protein response is an intricate system of sensor proteins in the endoplasmic reticulum (ER) that recognizes misfolded proteins and transmits information via transcription factors to either regain proteostasis or, depending on the severity, to induce apoptosis. The main transmembrane sensor is IRE1α, which contains cytoplasmic kinase and RNase domains relevant for its activation and the mRNA splicing of the transcription factor XBP1. Mast cell leukemia (MCL) is a severe form of systemic mastocytosis. The inhibition of IRE1α in the MCL cell line HMC-1.2 has anti-proliferative and pro-apoptotic effects, motivating us to elucidate the IRE1α interactors/regulators in HMC-1.2 cells. Therefore, the TurboID proximity labeling technique combined with MS analysis was applied. Gene Ontology and pathway enrichment analyses revealed that the majority of the enriched proteins are involved in vesicle-mediated transport, protein stabilization, and ubiquitin-dependent ER-associated protein degradation pathways. In particular, the AAA ATPase VCP and the oncoprotein MTDH as IRE1α-interacting proteins caught our interest for further analyses. The pharmacological inhibition of VCP activity resulted in the increased stability of IRE1α and MTDH as well as the activation of IRE1α. The interaction of VCP with both IRE1α and MTDH was dependent on ubiquitination. Moreover, MTDH stability was reduced in IRE1α-knockout cells. Hence, pharmacological manipulation of IRE1α-MTDH-VCP complex(es) might enable the treatment of MCL.
Collapse
Affiliation(s)
- Nabil Ahmed
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany (T.W.)
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany;
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany (T.W.)
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany (T.W.)
| |
Collapse
|
5
|
Polivka L, Madrange M, Bulai-Livideanu C, Barete S, Ballul T, Neuraz A, Greco C, Agopian J, Brenet F, Dubreuil P, Burdet C, Lemal R, Tournilhac O, Terriou L, Launay D, Bouillet L, Gourguechon C, Damaj G, Frenzel L, Meni C, Bouktit H, Collange AF, Gaudy-Marqueste C, Gousseff M, Le Mouel E, Hamidou M, Neel A, Ranta D, Jaussaud R, Guilpain P, Canioni D, Molina TJ, Bruneau J, Lhermitte L, Garcelon N, Javier RM, Pelletier F, Castelain F, Retornaz F, Cabrera Q, Zunic P, Gourin MP, Wierzbicka-Hainaut E, Viallard JF, Lavigne C, Hoarau C, Durieu I, Heiblig M, Dimicoli-Salazar S, Torregrosa-Diaz JM, Soria A, Arock M, Lortholary O, Bodemer C, Hermine O, Rossignol J. Pathophysiologic implications of elevated prevalence of hereditary alpha-tryptasemia in all mastocytosis subtypes. J Allergy Clin Immunol 2024; 153:349-353.e4. [PMID: 37633651 DOI: 10.1016/j.jaci.2023.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/22/2023] [Accepted: 08/08/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Mastocytosis and monoclonal mast cell (MC) activation syndrome (MMAS) are heterogeneous conditions characterized by the accumulation of atypical MCs. Despite the recurrent involvement of KIT mutations, the pathophysiologic origin of mastocytosis and MMAS is unclear. Although hereditary α-tryptasemia (HαT, related to TPSAB1 gene duplication) is abnormally frequent in these diseases, it is not known whether the association is coincidental or causal. OBJECTIVE We evaluated the prevalence of HαT in all mastocytosis subtypes and MMAS and assessed the pathophysiologic association with HαT. METHODS Clinical data, laboratory data, KIT mutations, TPSAB1 duplication (assessed by droplet digital PCR), and HαT prevalence were retrospectively recorded for all patients with mastocytosis and MMAS registered in the French national referral center database and compared to a control cohort. To increase the power of our analysis for advanced systemic mastocytosis (advSM), we pooled our cohort with literature cases. RESULTS We included 583 patients (27 with MMAS and 556 with mastocytosis). The prevalence of HαT in mastocytosis was 12.6%, significantly higher than in the general population (5.7%, P = .002) and lower than in MMAS (33.3%, P = .02). HαT+ patients were more likely to have anaphylactic reactions and less likely to have cutaneous lesions than HαT- patients (43.0% vs 24.4%, P = .006; 57.7% vs 75.6%, respectively, P = .006). In the pooled analysis, the prevalence of HαT was higher in advSM (11.5%) than in control cohorts (5.2%, P = .01). CONCLUSION Here we confirm the increase incidence of anaphylaxis in HαT+ mastocytosis patients. The increased prevalence of HαT in all subtypes of systemic mastocytosis (including advSM) is suggestive of pathophysiologic involvement.
Collapse
Affiliation(s)
- Laura Polivka
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | - Marine Madrange
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | | | - Stéphane Barete
- CEREMAST, the Department of Dermatology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Thomas Ballul
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Department of Hematology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| | - Antoine Neuraz
- Department of Bioinformatics, Necker-Children's Hospital, AP-HP, Paris Centre University, Imagine Institute, INSERM U1163, Paris, France
| | - Celine Greco
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Department of Pain and Palliative Care Unit, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| | - Julie Agopian
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France; Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytoses (AFIRMM), Marseille, France
| | - Fabienne Brenet
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France; Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytoses (AFIRMM), Marseille, France
| | - Patrice Dubreuil
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France; Association Française pour les Initiatives de Recherche sur le Mastocyte et les Mastocytoses (AFIRMM), Marseille, France
| | - Charles Burdet
- Centre d'Investigation Clinique, INSERM CIC 1425, AP-HP, Bichat Hospital, Paris Centre University, Paris, France
| | - Richard Lemal
- Histocompatibility Laboratory, EA 7453, Université Clermont Auvergne, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Olivier Tournilhac
- CEREMAST, the Adult Clinical Hematology, CHU Clermont-Ferrand, INSERM CIC501, EA 7453, Clermont Auvergne University, Clermont-Ferrand, France
| | - Louis Terriou
- CEREMAST, the Department of Internal Medicine and Clinical Immunology, Claude Huriez Hospital, CHRU Lille, Lille, France
| | - David Launay
- CEREMAST, the Department of Internal Medicine and Clinical Immunology, Claude Huriez Hospital, CHRU Lille, Lille, France; Lille University, INSERM U995 LIRIC, CHU Lille, and Referral Center for Rare Systemic Autoimmune Diseases North and North-west of France, Lille, France
| | - Laurence Bouillet
- CEREMAST, the Clinical Immunology/Internal Medicine Department, National Reference Center for Angioedema, Grenoble University Hospital, Grenoble, France
| | | | - Ghandi Damaj
- CEREMAST, the Haematology Institute, Normandy University School of Medicine, Caen, France
| | - Laurent Frenzel
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Department of Hematology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| | - Cécile Meni
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | - Hassiba Bouktit
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | - Anne Florence Collange
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | - Caroline Gaudy-Marqueste
- CEREMAST, the Department of Dermatology, Aix-Marseille University, CHU Timone, Marseille, France
| | - Marie Gousseff
- Department of Internal Medicine, Centre Hospitalier Bretagne Atlantique, Vannes, France
| | - Edwige Le Mouel
- CEREMAST, the Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | - Mohamed Hamidou
- CEREMAST, the Department of Internal Medicine, Hôtel-Dieu University Hospital, Nantes, France
| | - Antoine Neel
- CEREMAST, the Department of Internal Medicine, Hôtel-Dieu University Hospital, Nantes, France
| | - Dana Ranta
- Department of Haematology, Nancy University Hospital, Nancy, France
| | - Roland Jaussaud
- Department of Internal Medicine and Clinical Immunology, Vandoeuvre-lès-Nancy, France
| | - Philippe Guilpain
- CEREMAST, the Department of Internal Medicine-Multi-organ Diseases, Saint-Eloi University Hospital, Montpellier University, Montpellier, France
| | - Danielle Canioni
- CEREMAST, the Department of Pathology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| | - Thierry Jo Molina
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Department of Pathology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| | - Julie Bruneau
- CEREMAST, the Department of Pathology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| | - Ludovic Lhermitte
- CEREMAST, the Laboratory of Onco-hematology, Necker Children's Hospital, AP-HP, Paris, France
| | - Nicolas Garcelon
- Paris Centre University, Imagine Institute, Data Science Platform, INSERM UMR 1163, F-75015, Paris, France
| | - Rose-Marie Javier
- CEREMAST, the Department of Rheumatology, Strasbourg University Hospital, Strasbourg, France
| | - Fabien Pelletier
- CEREMAST, the Department of Dermatology, Allergology Unit, University Hospital of Besançon, Besançon, France
| | - Florence Castelain
- CEREMAST, the Department of Dermatology, Allergology Unit, University Hospital of Besançon, Besançon, France
| | - Frederique Retornaz
- Unité de soins et de recherche en médecine interne et maladies infectieuses, European Hospital, Marseille, France
| | - Quentin Cabrera
- Department of Haematology, Sud Reunion University Hospital, Saint Pierre, La Réunion, France
| | - Patricia Zunic
- Department of Haematology, Sud Reunion University Hospital, Saint Pierre, La Réunion, France
| | | | | | - Jean François Viallard
- Department of Internal Medicine and Infectious Diseases, Haut-Lévêque Hospital, CHRU Bordeaux, Bordeaux University, Bordeaux, France
| | - Christian Lavigne
- CEREMAST, the Department of Internal Medicine and Clinical Immunology, University Hospital, Angers, France
| | - Cyrille Hoarau
- CEREMAST, the Service d'Immunologie Clinique et d'Allergologie, Centre Hospitalier Régional Universitaire, Tours, France
| | - Isabelle Durieu
- CEREMAST, the Department of Internal Medicine, Adult Cystic Fibrosis Care Center, Hospices Civils de Lyon, Lyon, France
| | - Maël Heiblig
- CEREMAST, the Department of Hematology, Lyon-Sud Hospital, Hospices Civils de Lyon, Pierre-Bénite, France
| | | | | | - Angèle Soria
- CEREMAST, the Department of Dermatology and Allergy, Tenon Hospital, Sorbonne University, Paris, France
| | - Michel Arock
- CEREMAST, the Laboratory of Hematology, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Olivier Lortholary
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | - Christine Bodemer
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France
| | - Olivier Hermine
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Department of Hematology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France.
| | - Julien Rossignol
- CEREMAST, the Imagine Institute, INSERM U1163, AP-HP, Necker-Children's Hospital, Paris Centre University, Paris, France; CEREMAST, the Department of Hematology, Necker-Children's Hospital, AP-HP, Paris Centre University, Paris, France
| |
Collapse
|
6
|
Rajan V, Prykhozhij SV, Pandey A, Cohen AM, Rainey JK, Berman JN. KIT D816V is dimerization-independent and activates downstream pathways frequently perturbed in mastocytosis. Br J Haematol 2023; 202:960-970. [PMID: 35245395 DOI: 10.1111/bjh.18116] [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: 12/12/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
KIT, a type III tyrosine kinase receptor, plays a crucial role in haematopoietic development. The KIT receptor forms a dimer after ligand binding; this activates tyrosine kinase activity leading to downstream signal transduction. The D816V KIT mutation is extensively implicated in haematological malignancies, including mastocytosis and leukaemia. KIT D816V is constitutively active, but the molecular nuances that lead to constitutive tyrosine kinase activity are unclear. For the first time, we present experimental evidence that the KIT D816V mutant does not dimerize like KIT wild type. We further show evidence of decreased stabilization of the tyrosine kinase domain in the KIT D816V mutant, a phenomenon that might contribute to its constitutive activity. Since the mechanism of KIT D816V activation varies from that of the wild type, we explored downstream signal transduction events and found that even though KIT D816V targets similar signalling moieties, the signalling is amplified in the mutant compared to stem cell factor-activated wild type receptor. Uniquely, KIT D816V induces infection-related pathways and the spliceosome pathway, providing alternate options for selective as well as combinatorial therapeutic targeting.
Collapse
Affiliation(s)
- Vinothkumar Rajan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Sergey V Prykhozhij
- Children's Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, ON, Canada
| | - Aditya Pandey
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Alejandro M Cohen
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Jan K Rainey
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, Canada
- Department of Chemistry, Dalhousie University, Halifax, NS, Canada
- School of Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
| | - Jason N Berman
- Children's Hospital of Eastern Ontario (CHEO) Research Institute and University of Ottawa, Ottawa, ON, Canada
- Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| |
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: 30] [Impact Index Per Article: 15.0] [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
|
Carter MC, Park J, Vadas P, Worm M. Extrinsic and Intrinsic Modulators of Anaphylaxis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1998-2006. [PMID: 37230384 DOI: 10.1016/j.jaip.2023.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
The severity of anaphylaxis is determined by many factors. The allergenic source as well as the age of the affected individual and the route of allergen exposure encompass the major contributors of the clinical outcome. Moreover, the severity can be modulated further by intrinsic and extrinsic factors. Among these, the genetic predisposition, certain comorbidities such as uncontrolled asthma, and hormonal fluctuations have been proposed as intrinsic and antihypertensive medications or physical activity as extrinsic factors. Recent advances have highlighted immunologic pathways that may exacerbate the response to allergens through receptors on mast cells, basophils, platelets, and other granulocytes. Atopy, platelet-activating factor acetylhydrolase deficiency, hereditary alpha tryptasemia, and clonal mast cell disorders are examples associated with genetic alterations that may predispose to severe anaphylaxis. Identifying risk factors that lower the threshold of reactivity or increase the severity of multisystem reactions is important in the management of this patient population.
Collapse
Affiliation(s)
- Melody C Carter
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Jane Park
- Division of Clinical Immunology and Allergy, Department of Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Peter Vadas
- Division of Clinical Immunology and Allergy, Department of Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Margitta Worm
- Division of Allergy and Immunology, Department of Dermatology, Venerology and Allergy, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
9
|
Kaszuba A, Sławińska M, Żółkiewicz J, Sobjanek M, Nowicki RJ, Lange M. Mastocytosis and Skin Cancer: The Current State of Knowledge. Int J Mol Sci 2023; 24:9840. [PMID: 37372988 DOI: 10.3390/ijms24129840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/26/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Mastocytosis is a heterogeneous group of diseases associated with excessive proliferation and accumulation of mast cells in different organs. Recent studies have demonstrated that patients suffering from mastocytosis face an increased risk of melanoma and non-melanoma skin cancer. The cause of this has not yet been clearly identified. In the literature, the potential influence of several factors has been suggested, including genetic background, the role of cytokines produced by mast cells, iatrogenic and hormonal factors. The article summarizes the current state of knowledge regarding the epidemiology, pathogenesis, diagnosis, and management of skin neoplasia in mastocytosis patients.
Collapse
Affiliation(s)
- Agnieszka Kaszuba
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Smoluchowskiego Street 17, 80-214 Gdańsk, Poland
| | - Martyna Sławińska
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Smoluchowskiego Street 17, 80-214 Gdańsk, Poland
| | - Jakub Żółkiewicz
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Smoluchowskiego Street 17, 80-214 Gdańsk, Poland
| | - Michał Sobjanek
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Smoluchowskiego Street 17, 80-214 Gdańsk, Poland
| | - Roman J Nowicki
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Smoluchowskiego Street 17, 80-214 Gdańsk, Poland
| | - Magdalena Lange
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, Smoluchowskiego Street 17, 80-214 Gdańsk, Poland
| |
Collapse
|
10
|
Murray HC, Miller K, Brzozowski JS, Kahl RGS, Smith ND, Humphrey SJ, Dun MD, Verrills NM. Synergistic Targeting of DNA-PK and KIT Signaling Pathways in KIT Mutant Acute Myeloid Leukemia. Mol Cell Proteomics 2023; 22:100503. [PMID: 36682716 PMCID: PMC9986649 DOI: 10.1016/j.mcpro.2023.100503] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 12/19/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common and aggressive form of acute leukemia, with a 5-year survival rate of just 24%. Over a third of all AML patients harbor activating mutations in kinases, such as the receptor tyrosine kinases FLT3 (receptor-type tyrosine-protein kinase FLT3) and KIT (mast/stem cell growth factor receptor kit). FLT3 and KIT mutations are associated with poor clinical outcomes and lower remission rates in response to standard-of-care chemotherapy. We have recently identified that the core kinase of the non-homologous end joining DNA repair pathway, DNA-PK (DNA-dependent protein kinase), is activated downstream of FLT3; and targeting DNA-PK sensitized FLT3-mutant AML cells to standard-of-care therapies. Herein, we investigated DNA-PK as a possible therapeutic vulnerability in KIT mutant AML, using isogenic FDC-P1 mouse myeloid progenitor cell lines transduced with oncogenic mutant KIT (V560G and D816V) or vector control. Targeted quantitative phosphoproteomic profiling identified phosphorylation of DNA-PK in the T2599/T2605/S2608/S2610 cluster in KIT mutant cells, indicative of DNA-PK activation. Accordingly, proliferation assays revealed that KIT mutant FDC-P1 cells were more sensitive to the DNA-PK inhibitors M3814 or NU7441, compared with empty vector controls. DNA-PK inhibition combined with inhibition of KIT signaling using the kinase inhibitors dasatinib or ibrutinib, or the protein phosphatase 2A activators FTY720 or AAL(S), led to synergistic cell death. Global phosphoproteomic analysis of KIT-D816V cells revealed that dasatinib and M3814 single-agent treatments inhibited extracellular signal-regulated kinase and AKT (RAC-alpha serine/threonine-protein kinase)/MTOR (serine/threonine-protein kinase mTOR) activity, with greater inhibition of both pathways when used in combination. Combined dasatinib and M3814 treatment also synergistically inhibited phosphorylation of the transcriptional regulators MYC and MYB. This study provides insight into the oncogenic pathways regulated by DNA-PK beyond its canonical role in DNA repair and demonstrates that DNA-PK is a promising therapeutic target for KIT mutant cancers.
Collapse
Affiliation(s)
- Heather C Murray
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Kasey Miller
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Joshua S Brzozowski
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Richard G S Kahl
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Nathan D Smith
- Analytical and Biomolecular Research Facility, Advanced Mass Spectrometry Unit, University of Newcastle, Callaghan, New South Wales, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, and The Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew D Dun
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, and Hunter Cancer Research Alliance and Precision Medicine Program, Hunter Medical Research Institute, Callaghan, New South Wales, Australia.
| |
Collapse
|
11
|
Pfeiffer A, Petersen JD, Falduto GH, Anderson DE, Zimmerberg J, Metcalfe DD, Olivera A. Selective immunocapture reveals neoplastic human mast cells secrete distinct microvesicle- and exosome-like populations of KIT-containing extracellular vesicles. J Extracell Vesicles 2022; 11:e12272. [PMID: 36239715 PMCID: PMC9838129 DOI: 10.1002/jev2.12272] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 08/03/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
Activating mutations in the receptor KIT promote the dysregulated proliferation of human mast cells (huMCs). The resulting neoplastic huMCs secrete extracellular vesicles (EVs) that can transfer oncogenic KIT among other cargo into recipient cells. Despite potential contributions to diseases, KIT-containing EVs have not been thoroughly investigated. Here, we isolated and characterized KIT-EV subpopulations released by neoplastic huMCs using an immunocapture approach that selectively isolates EVs containing KIT in its proper topology. Immunocapture of EVs on KIT antibody-coated electron microscopy (EM) affinity grids allowed to assess the morphology and size of KIT-EVs. Immunoblot analysis demonstrated KIT-EVs have a distinct protein profile from KIT-depleted EVs, contain exosome and microvesicle markers, and are separated into these subtypes by ultracentrifugation. Cell treatment with sphingomyelinase inhibitors shifted the protein content among KIT-EV subtypes, suggesting different biogenesis routes. Proteomic analysis revealed huMC KIT-EVs are enriched in proteins involved in signalling, immune responses, and cell migration, suggesting diverse biological functions, and indicated neoplastic huMCs disseminate KIT via shuttling in heterogeneous microvesicle- and exosome-like EVs. Further, selective KIT-immunocapture will enable the enrichment of specific huMC-derived EVs from complex human biosamples and facilitate an understanding of their in vivo functions and potential to serve as biomarkers of specific biological pathologies.
Collapse
Affiliation(s)
- Annika Pfeiffer
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Jennifer D. Petersen
- Section on Integrative BiophysicsDivision of Basic and Translational BiophysicsEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Guido H. Falduto
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - David Eric Anderson
- Advanced Mass Spectrometry Core FacilityNational Institute of Diabetes and Digestive and Kidney DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Joshua Zimmerberg
- Section on Integrative BiophysicsDivision of Basic and Translational BiophysicsEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentNational Institutes of HealthBethesdaMarylandUSA
| | - Dean D. Metcalfe
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| | - Ana Olivera
- Mast Cell Biology SectionLaboratory of Allergic DiseasesNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
12
|
Hasegawa Y, Shosu K, Tsuji K, Shimoyama Y, Miyama TS, Baba K, Okuda M, Itamoto K, Igase M, Mizuno T. Intratumoral heterogeneity of c-KIT mutations in a feline splenic mast cell tumor and their functional effects on cell proliferation. Sci Rep 2022; 12:15791. [PMID: 36138037 PMCID: PMC9499958 DOI: 10.1038/s41598-022-19089-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
A cat was presented with mast cell tumors (MCTs) of the skin and spleen. During the initial diagnosis, the exon 8 mutation of c-KIT was detected in the masses from skin and spleen by a commercial laboratory test. Consequently, treatment with toceranib was started. After complete remission, because of recurrence on day 117, the spleen and skin tumors were removed, but the cat eventually died on day 191. The analysis of ten cDNA clones of the c-KIT gene cloned from the surgically removed spleen revealed that seven different cDNA patterns were included, indicating the heterogeneity of this gene in the splenic MCT. The seven cDNA nucleotide patterns can be classified into four protein sequence patterns. In addition to the previously known mutations in exon 8, we identified novel mutations in exons 9, 10, and 18; four amino acids deletion in exon 9, and a point mutation in exons 10 and 18. Mouse IL-3-dependent cell line, Ba/F3, was transduced with these mutant clones, and c-KIT phosphorylation and proliferation assays were performed. We found that certain mutations affected the c-KIT phosphorylation status and cell proliferation. This suggests that heterogeneity among the population of tumor cells exists in MCTs, and that the dominant clones of this heterogeneity may contribute to the subsequent tumor cell growth.
Collapse
Affiliation(s)
- Yuki Hasegawa
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kazuha Shosu
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kanako Tsuji
- Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | | | - Takako Shimokawa Miyama
- Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kenji Baba
- Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masaru Okuda
- Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kazuhito Itamoto
- Laboratory of Small Animal Clinical Science, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masaya Igase
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takuya Mizuno
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
13
|
De Vos S, Demeyere K, De Cock H, Devriendt N, Schwarzkopf I, Fortrie R, Roggeman T, Meyer E, De Spiegelaere W, de Rooster H. Comparison of serum tryptase as a diagnostic oncological marker in canine versus human mast cell neoplasms. Res Vet Sci 2022; 151:90-95. [DOI: 10.1016/j.rvsc.2022.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 04/20/2022] [Accepted: 05/02/2022] [Indexed: 11/24/2022]
|
14
|
González-López O, Muñoz-González JI, Orfao A, Álvarez-Twose I, García-Montero AC. Comprehensive Analysis of Acquired Genetic Variants and Their Prognostic Impact in Systemic Mastocytosis. Cancers (Basel) 2022; 14:cancers14102487. [PMID: 35626091 PMCID: PMC9139197 DOI: 10.3390/cancers14102487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 01/27/2023] Open
Abstract
Systemic mastocytosis (SM) is a rare clonal haematopoietic stem cell disease in which activating KIT mutations (most commonly KIT D816V) are present in virtually every (>90%) adult patient at similar frequencies among non-advanced and advanced forms of SM. The KIT D816V mutation is considered the most common pathogenic driver of SM. Acquisition of this mutation early during haematopoiesis may cause multilineage involvement of haematopoiesis by KIT D816V, which has been associated with higher tumour burden and additional mutations in other genes, leading to an increased rate of transformation to advanced SM. Thus, among other mutations, alterations in around 30 genes that are also frequently mutated in other myeloid neoplasms have been reported in SM cases. From these genes, 12 (i.e., ASXL1, CBL, DNMT3A, EZH2, JAK2, KRAS, NRAS, SF3B1, RUNX1, SF3B1, SRSF2, TET2) have been recurrently reported to be mutated in SM. Because of all the above, assessment of multilineage involvement of haematopoiesis by the KIT D816V mutation, in the setting of multi-mutated haematopoiesis as revealed by a limited panel of genes (i.e., ASXL1, CBL, DNMT3A, EZH2, NRAS, RUNX1 and SRSF2) and associated with a poorer patient outcome, has become of great help to identify SM patients at higher risk of disease progression and/or poor survival who could benefit from closer follow-up and eventually also early cytoreductive treatment.
Collapse
Affiliation(s)
- Oscar González-López
- Cancer Research Center (IBMCC, USAL/CSIC), Department of Medicine, Universidad de Salamanca, Biomedical Research Institute of Salamanca and Spanish Network on Mastocytosis (REMA), 37007 Salamanca, Spain; (O.G.-L.); (J.I.M.-G.); (A.O.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Javier I. Muñoz-González
- Cancer Research Center (IBMCC, USAL/CSIC), Department of Medicine, Universidad de Salamanca, Biomedical Research Institute of Salamanca and Spanish Network on Mastocytosis (REMA), 37007 Salamanca, Spain; (O.G.-L.); (J.I.M.-G.); (A.O.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Alberto Orfao
- Cancer Research Center (IBMCC, USAL/CSIC), Department of Medicine, Universidad de Salamanca, Biomedical Research Institute of Salamanca and Spanish Network on Mastocytosis (REMA), 37007 Salamanca, Spain; (O.G.-L.); (J.I.M.-G.); (A.O.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Iván Álvarez-Twose
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast, Virgen del Valle Hospital) and REMA, 45071 Toledo, Spain
| | - Andrés C. García-Montero
- Cancer Research Center (IBMCC, USAL/CSIC), Department of Medicine, Universidad de Salamanca, Biomedical Research Institute of Salamanca and Spanish Network on Mastocytosis (REMA), 37007 Salamanca, Spain; (O.G.-L.); (J.I.M.-G.); (A.O.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Correspondence:
| |
Collapse
|
15
|
Snider DB, Arthur GK, Falduto GH, Olivera A, Ehrhardt-Humbert LC, Smith E, Smith C, Metcalfe DD, Cruse G. Targeting KIT by frameshifting mRNA transcripts as a therapeutic strategy for aggressive mast cell neoplasms. Mol Ther 2022; 30:295-310. [PMID: 34371183 PMCID: PMC8753370 DOI: 10.1016/j.ymthe.2021.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/21/2021] [Accepted: 07/31/2021] [Indexed: 01/07/2023] Open
Abstract
Activating mutations in c-KIT are associated with the mast cell (MC) clonal disorders cutaneous mastocytosis and systemic mastocytosis and its variants, including aggressive systemic mastocytosis, MC leukemia, and MC sarcoma. Currently, therapies inhibiting KIT signaling are a leading strategy to treat MC proliferative disorders. However, these approaches may have off-target effects, and in some patients, complete remission or improved survival time cannot be achieved. These limitations led us to develop an approach using chemically stable exon skipping oligonucleotides (ESOs) that induce exon skipping of precursor (pre-)mRNA to alter gene splicing and introduce a frameshift into mature KIT mRNA transcripts. The result of this alternate approach results in marked downregulation of KIT expression, diminished KIT signaling, inhibition of MC proliferation, and rapid induction of apoptosis in neoplastic HMC-1.2 MCs. We demonstrate that in vivo administration of KIT targeting ESOs significantly inhibits tumor growth and systemic organ infiltration using both an allograft mastocytosis model and a humanized xenograft MC tumor model. We propose that our innovative approach, which employs well-tolerated, chemically stable oligonucleotides to target KIT expression through unconventional pathways, has potential as a KIT-targeted therapeutic alone, or in combination with agents that target KIT signaling, in the treatment of KIT-associated malignancies.
Collapse
Affiliation(s)
- Douglas B. Snider
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA,Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| | - Greer K. Arthur
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA,Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA
| | - Guido H. Falduto
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ana Olivera
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lauren C. Ehrhardt-Humbert
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA
| | - Emmaline Smith
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA
| | - Cierra Smith
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA
| | - Dean D. Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Glenn Cruse
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA,Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27607, USA,Corresponding author: Glenn Cruse, PhD, Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Biomedical Partnership Center, 1060 William Moore Drive, Raleigh, NC 27607, USA.
| |
Collapse
|
16
|
Polivka L, Parietti V, Bruneau J, Soucie E, Madrange M, Bayard E, Rignault R, Canioni D, Fraitag S, Lhermitte L, Feroul M, Tissandier M, Rossignol J, Frenzel L, Cagnard N, Meni C, Bouktit H, Collange AF, Gougoula C, Parisot M, Bader-Meunier B, Livideanu C, Laurent C, Arock M, Hadj-Rabia S, Rüther U, Dubreuil P, Bodemer C, Hermine O, Maouche-Chrétien L. The association of Greig syndrome and mastocytosis reveals the involvement of the hedgehog pathway in advanced mastocytosis. Blood 2021; 138:2396-2407. [PMID: 34424959 DOI: 10.1182/blood.2020010207] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/28/2021] [Indexed: 11/20/2022] Open
Abstract
Mastocytosis is a heterogeneous disease characterized by an abnormal accumulation of mast cells (MCs) in 1 or several organs. Although a somatic KIT D816V mutation is detected in ∼85% of patients, attempts to demonstrate its oncogenic effect alone have repeatedly failed, suggesting that additional pathways are involved in MC transformation. From 3 children presenting with both Greig cephalopolysyndactyly syndrome (GCPS, Mendelian Inheritance in Man [175700]) and congenital mastocytosis, we demonstrated the involvement of the hedgehog (Hh) pathway in mastocytosis. GCPS is an extremely rare syndrome resulting from haploinsufficiency of GLI3, the major repressor of Hh family members. From these familial cases of mastocytosis, we demonstrate that the Hh pathway is barely active in normal primary MCs and is overactive in neoplastic MCs. GLI3 and KIT mutations had a synergistic, tumorigenic effect on the onset of mastocytosis in a GCPS mouse model. Finally, Hh inhibitors suppressed neoplastic MC proliferation in vitro and extend the survival time of mice with aggressive systemic mastocytosis (ASM). This work revealed, for the first time, the involvement of Hh signaling in the pathophysiology of mastocytosis and demonstrated the cooperative effects of the KIT and Hh oncogenic pathways in mice with ASM, leading to the identification of new promising therapeutic targets.
Collapse
Affiliation(s)
- L Polivka
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - V Parietti
- Department of Animal Experimentation, Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - J Bruneau
- Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris-Centre University, Paris, France
| | - E Soucie
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France
| | - M Madrange
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - E Bayard
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - R Rignault
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - D Canioni
- Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris-Centre University, Paris, France
| | - S Fraitag
- Department of Pathology, Hôpital Necker-Enfants Malades, AP-HP, Paris-Centre University, Paris, France
| | - L Lhermitte
- Institut Necker-Enfants Malades, Université de Paris, INSERM Unité (U)1151, Paris, France
- Laboratory of Onco-Hematology, Hôpital Universitaire Necker Enfants-Malades, AP-HP, Paris, France
| | - M Feroul
- Institut Necker-Enfants Malades, Université de Paris, INSERM Unité (U)1151, Paris, France
- Laboratory of Onco-Hematology, Hôpital Universitaire Necker Enfants-Malades, AP-HP, Paris, France
| | - M Tissandier
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
| | - J Rossignol
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - L Frenzel
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
- Department of Hematology, Necker-Enfants Malades Hospital, AP-HP, Paris-Centre University, Imagine Institute, Paris, France
| | - N Cagnard
- Bioinformatics, Platform Bioinformatics, INSERM U1163, Paris-Centre University, Imagine Institute, Paris, France
| | - C Meni
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - H Bouktit
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - A-F Collange
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - C Gougoula
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT), Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - M Parisot
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163 et INSERM US24/Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Service (UMS)3633, Paris-Centre University, Imagine Institute, Paris, France
| | - B Bader-Meunier
- Department of Pediatric Immunology and Hematology, Necker-Enfants Malades Hospital, AP-HP, INSERM U1163, Paris-Centre University, Paris, France
| | - C Livideanu
- Service de Dermatologie, CEREMAST, CHU de Toulouse
| | - C Laurent
- Service d'Anatomie-Pathologique, Oncopole, Centre Hospitalier de Universitaire (CJU) de Toulouse, Toulouse, France
| | - M Arock
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
- Centre de Recherche des Cordeliers, INSERM Unité de Recherche Mixte en Santé (UMRS)1138, Paris, France
- Laboratory of Hematology, Pitié-Salpêtrière Hospital, AP-HP Sorbonne Université, Paris, France; and
| | - S Hadj-Rabia
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - U Rüther
- Institute of Animal Developmental and Molecular Biology, Heinrich Heine University, Düsseldorf, Germany
| | - P Dubreuil
- Centre de Recherche en Cancérologie de Marseille, INSERM U1068, Marseille, France
| | - C Bodemer
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| | - O Hermine
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
- Department of Hematology, Necker-Enfants Malades Hospital, AP-HP, Paris-Centre University, Imagine Institute, Paris, France
| | - L Maouche-Chrétien
- Laboratory of Molecular Mechanisms of Hematologic Disorders and Therapeutic Implications, INSERM Unité Mixte de Recherche (UMR) 1163, Paris-Centre University, Imagine Institute, Paris, France
- Department of Dermatology, Reference Center for Genodermatoses (MAGEC), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Centre de Référence Maladies Rares des Mastocytoses (CEREMAST), Necker-Enfants Malades Hospital, Paris, France
| |
Collapse
|
17
|
Sumbly V, Landry I, Iqbal S, Bhatti Z, Alshamam MS, Ashfaq S, Rizzo V. The Role of Avapritinib for the Treatment of Systemic Mastocytosis. Cureus 2021; 13:e18385. [PMID: 34729266 PMCID: PMC8556140 DOI: 10.7759/cureus.18385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
Systemic mastocytosis is a rare hematologic disorder characterized by the clonal proliferation of mast cells in extra-cutaneous organs. This disease can be further subdivided into five different phenotypes: indolent systemic mastocytosis (ISM), smoldering systemic mastocytosis (SSM), aggressive systemic mastocytosis (ASM), systemic mastocytosis with an associated hematological neoplasm (SM-AHN) and mast cell leukemia (MCL). The tyrosine kinase inhibitor (and also potent KIT D816V inhibitor) avapritinib, initially approved for the treatment of gastrointestinal stromal tumors (GISTs) bearing a PDGFRA exon 18 mutation, also showed great promise in patients with systemic mastocytosis, a disease known to be driven by a mutation in KIT (D816V). We present an overview of this rare disorder, including a review of the current understanding of the genetic mechanisms which lead to the disease state, the action of the tyrosine kinase inhibitors, as well as the latest clinical trial data which led to the current recommendations for the use of avapritinib.
Collapse
Affiliation(s)
- Vikram Sumbly
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals, Queens, USA
| | - Ian Landry
- Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals, Queens, USA
| | - Saba Iqbal
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals, Queens, USA
| | - Zamaraq Bhatti
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals, Queens, USA
| | - Mohsen S Alshamam
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals, Queens, USA
| | | | - Vincent Rizzo
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals, Queens, USA
| |
Collapse
|
18
|
PATHOGENIC AND DIAGNOSTIC RELEVANCE OF KIT IN PRIMARY MAST CELL ACTIVATION DISORDERS. Ann Allergy Asthma Immunol 2021; 127:427-434. [PMID: 34298172 DOI: 10.1016/j.anai.2021.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/17/2021] [Accepted: 07/15/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Mast cell (MC) activation (MCA) defines the mechanism by which certain patients suffer from symptoms due to the effect of a wide range of mediators released from MC upon their activation triggered by different stimuli. When these symptoms are severe and recurrent, the diagnosis of MCA syndrome (MCAS) might be considered. Here we review the relevant aspects related to the pathogenesis of MCAS, with special emphasis on the prevalence and diagnostic relevance of KIT mutations. DATA SOURCES PubMed was searched between 1980 and 2021 using the following terms: Mast cell activation syndromes, mast cell activation, anaphylaxis, KIT mutations, KIT D816V, indolent systemic mastocytosis, bone marrow mastocytosis, cutaneous mastocytosis, IgE anaphylaxis and idiopathic anaphylaxis. STUDY SELECTIONS Only articles published in English were selected based on their relevance to MCAS and/or severe and recurrent anaphylaxis. RESULTS MCAS can be classified in clonal MCAS and non-clonal MCAS depending on the presence vs. absence of an underlying KIT mutation (mostly KIT D816V), respectively. In contrast to clonal MCAS in which MCA is associated with a primary MC disorder (i.e. primary MCAS) such as mastocytosis or monoclonal MCAS, non-clonal MCAS can be secondary to known or unidentified triggers (i.e. secondary and idiopathic MCAS, respectively). CONCLUSION The clinical heterogeneity and complexity of the molecular assays needed for the study of MCAS patients might lead to misdiagnosis, particularly when patients are evaluated at non-specialized centers. Thus, referral of patients suffering from clinical manifestations suggestive of MCAS to Reference Centers on mastocytosis and MC diseases is strongly recommended.
Collapse
|
19
|
Klug LR, Corless CL, Heinrich MC. Inhibition of KIT Tyrosine Kinase Activity: Two Decades After the First Approval. J Clin Oncol 2021; 39:1674-1686. [PMID: 33797935 DOI: 10.1200/jco.20.03245] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lillian R Klug
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR.,VA Portland Health Care System, Portland, OR
| | - Christopher L Corless
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Department of Pathology, Oregon Health & Science University, Portland, OR
| | - Michael C Heinrich
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR.,Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR.,VA Portland Health Care System, Portland, OR
| |
Collapse
|
20
|
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
|
21
|
Protein tyrosine phosphatase receptor type E (PTPRE) regulates the activation of wild-type KIT and KIT mutants differently. Biochem Biophys Rep 2021; 26:100974. [PMID: 33732906 PMCID: PMC7937656 DOI: 10.1016/j.bbrep.2021.100974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 11/20/2022] Open
Abstract
Activation of receptor tyrosine kinases needs tight control by tyrosine phosphatases to keep their normal function. In this study, we investigated the regulation of activation of the type III receptor tyrosine kinase KIT by protein tyrosine phosphatase receptor type E (PTPRE). We found that PTPRE can associate with wild-type KIT and inhibit KIT activation in a dose-dependent manner, although the activation of wild-type KIT is dramatically inhibited even when PTPRE is expressed at low level. The D816V mutation of KIT is the most frequently found oncogenic mutation in mastocytosis, and we found that PTPRE can associate and inhibit the activation of KIT/D816V in a dose dependent manner, but the inhibition is much weaker compared with wild-type KIT. Similar to mastocytosis, KIT mutations are the main oncogenic mutations in gastrointestinal stromal tumors (GISTs) although GISTs carry different types of KIT mutations. We further studied the regulation of the activation of GISTs-type KIT mutants and other mastocytosis-type KIT mutants by PTPRE. Indeed, PTPRE can almost block the activation of GISTs-type KIT mutants, while the activation of mastocytosis-type KIT mutants is more resistant to the inhibition of PTPRE. Taken together, our results suggest that PTPRE can associate with KIT, and inhibit the activation of both wild-type KIT and GISTs-type KIT mutants, while the activation of mastocytosis-type KIT mutants is more resistant to PTPRE. PTPRE associates with wild-type KIT and KIT mutants. PTPRE inhibits the activation of both wild-type KIT and GISTs-type KIT mutants dramatically. The activation of mastocytosis-type KIT mutants are more resistant to the inhibition of PTPRE.
Collapse
|
22
|
Lange M, Hartmann K, Carter MC, Siebenhaar F, Alvarez-Twose I, Torrado I, Brockow K, Renke J, Irga-Jaworska N, Plata-Nazar K, Ługowska-Umer H, Czarny J, Belloni Fortina A, Caroppo F, Nowicki RJ, Nedoszytko B, Niedoszytko M, Valent P. Molecular Background, Clinical Features and Management of Pediatric Mastocytosis: Status 2021. Int J Mol Sci 2021; 22:2586. [PMID: 33806685 PMCID: PMC7961542 DOI: 10.3390/ijms22052586] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022] Open
Abstract
Pediatric mastocytosis is a heterogeneous disease characterized by accumulation of mast cells in the skin and less frequently in other organs. Somatic or germline mutations in the KIT proto-oncogene are detected in most patients. Cutaneous mastocytosis is the most common form of the disease in children. In the majority of cases, skin lesions regress spontaneously around puberty. However, in few patients, mastocytosis is not a self-limiting disease, but persists into adulthood and can show signs of systemic involvement, especially when skin lesions are small-sized and monomorphic. Children with mastocytosis often suffer from mast cell mediator-related symptoms. Severe hypersensitivity reactions can also occur, mostly in patients with extensive skin lesions and blistering. In a substantial number of these cases, the triggering factor of anaphylaxis remains unidentified. Management of pediatric mastocytosis is mainly based on strict avoidance of triggers, treatment with H1 and H2 histamine receptor blockers, and equipment of patients and their families with epinephrine auto-injectors for use in severe anaphylactic reactions. Advanced systemic mastocytosis occurs occasionally. All children with mastocytosis require follow-up examinations. A bone marrow investigation is performed when advanced systemic mastocytosis is suspected and has an impact on therapy or when cutaneous disease persists into adulthood.
Collapse
Affiliation(s)
- Magdalena Lange
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (H.Ł.-U.); (J.C.); (R.J.N.); (B.N.)
| | - Karin Hartmann
- Division of Allergy, Department of Dermatology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland;
- Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Melody C. Carter
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Frank Siebenhaar
- Dermatological Allergology, Department of Dermatology and Allergy, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 10117 Berlin, Germany;
| | - Ivan Alvarez-Twose
- Instituto de Estudios de Mastocitosis de Castilla-La Mancha (CLMast)—Spanish Reference Center for Mastocytosis, Hospital Virgen del Valle—Complejo Hospitalario de Toledo, 45071 Toledo, Spain; (I.A.-T.); (I.T.)
| | - Inés Torrado
- Instituto de Estudios de Mastocitosis de Castilla-La Mancha (CLMast)—Spanish Reference Center for Mastocytosis, Hospital Virgen del Valle—Complejo Hospitalario de Toledo, 45071 Toledo, Spain; (I.A.-T.); (I.T.)
| | - Knut Brockow
- Department of Dermatology and Allergy Biederstein, School of Medicine, Technical University of Munich, 80802 Munich, Germany;
| | - Joanna Renke
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.R.); (N.I.-J.)
| | - Ninela Irga-Jaworska
- Department of Pediatrics, Hematology and Oncology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (J.R.); (N.I.-J.)
| | - Katarzyna Plata-Nazar
- Department of Pediatrics, Pediatric Gastroenterology, Allergology and Nutrition, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
| | - Hanna Ługowska-Umer
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (H.Ł.-U.); (J.C.); (R.J.N.); (B.N.)
| | - Justyna Czarny
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (H.Ł.-U.); (J.C.); (R.J.N.); (B.N.)
| | - Anna Belloni Fortina
- Pediatric Dermatology Unit, Department of Medicine DIMED, University of Padua, 35128 Padua, Italy; (A.B.F.); (F.C.)
| | - Francesca Caroppo
- Pediatric Dermatology Unit, Department of Medicine DIMED, University of Padua, 35128 Padua, Italy; (A.B.F.); (F.C.)
| | - Roman J. Nowicki
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (H.Ł.-U.); (J.C.); (R.J.N.); (B.N.)
| | - Bogusław Nedoszytko
- Department of Dermatology, Venereology and Allergology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (H.Ł.-U.); (J.C.); (R.J.N.); (B.N.)
| | - Marek Niedoszytko
- Department of Allergology, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
- Ludwig Boltzmann Institute for Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
23
|
Arun VA, Soni D, Bal A, Jain A. Aggressive systemic mastocytosis presenting as rapidly progressive ascites, generalised lymphadenopathy and osteosclerosis. BMJ Case Rep 2021; 14:14/2/e238034. [PMID: 33558379 PMCID: PMC7872913 DOI: 10.1136/bcr-2020-238034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Rapidly progressive ascites is a frequent clinical manifestation of advanced abdominal malignancies or portal hypertension due to liver diseases. We report a case of 61-year-old man who presented with rapidly progressive ascites. The presence of ascites, generalised lymphadenopathy, osteosclerosis on imaging and hepatosplenomegaly initially pointed towards the diagnosis of advanced high-grade lymphoma or accelerated myeloid neoplasm. Lymph node biopsy revealed infiltration by CD45, cKIT and CD30; tryptase and toluidine blue-positive mast cells (MCs). Bone marrow examination revealed infiltration by MCs and next generation sequencing revealed the pathognomic exon 17 D 816V KIT mutation. The patient was started on weekly pegylated interferon with significant symptom relief. Systemic mastocytosis should be considered as a differential diagnosis in a clinical case of ascites of unknown aetiology even in the absence of typical skin manifestations.
Collapse
Affiliation(s)
| | - Dipesh Soni
- Department of Internal Medicine, PGIMER, Chandigarh, India
| | - Amanjit Bal
- Department of Histopathology, PGIMER, Chandigarh, India
| | - Arihant Jain
- Department of Internal Medicine, PGIMER, Chandigarh, India
| |
Collapse
|
24
|
Mast Cell Activation Disorders. ACTA ACUST UNITED AC 2021; 57:medicina57020124. [PMID: 33573161 PMCID: PMC7911219 DOI: 10.3390/medicina57020124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/23/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023]
Abstract
Background and Objectives: Mast cell disorders comprise a wide spectrum of syndromes caused by mast cells' degranulation with acute or chronic clinical manifestations. Materials and Methods: In this review article we reviewed the latest findings in scientific papers about mast cell disorders with a particular focus on mast cell activation syndrome and mastocytosis in pediatric age. Results: Patients with mast cell activation syndrome have a normal number of mast cells that are hyperreactive upon stimulation of various triggers. We tried to emphasize the diagnostic criteria, differential diagnosis, and therapeutic strategies. Another primary mast cell disorder is mastocytosis, a condition with a long-known disease, in which patients have an increased number of mast cells that accumulate in different regions of the body with different clinical evolution in pediatric age. Conclusions: Mast cell activation syndrome overlaps with different clinical entities. No consensus was found on biomarkers and no clearly resolutive treatment is available. Therefore, a more detailed knowledge of this syndrome is of fundamental importance for a correct diagnosis and effective therapy.
Collapse
|
25
|
Criteria for the Regression of Pediatric Mastocytosis: A Long-Term Follow-Up. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 9:1695-1704.e5. [PMID: 33338682 DOI: 10.1016/j.jaip.2020.12.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Mastocytosis is a neoplastic condition characterized by the accumulation of mast cells (MCs) in 1 or more organ. Adults tend to have persistent, systemic mastocytosis, whereas MC infiltration in children is usually limited to the skin and typically regresses after several years. Both adults and children could display mast cell activation symptoms (MCASs) due to MC mediator release. In more than 85% of both adult and pediatric cases, KIT mutations are present, with the KIT D816V mutation being present in most affected adults but in only half the affected children. OBJECTIVE To identify the clinical, biological, and molecular factors associated with the regression of cutaneous mastocytosis (CM) in children, and to assess the correlation between MCASs and CM regression. METHODS Patients having suffered from pediatric-onset mastocytosis for at least 8 years were included in a longitudinal cohort study. Clinical data, the baseline serum tryptase level, the KIT sequence, and the progression of MCASs and CM were recorded. RESULTS CM regressed in 210 of the 272 included patients (77.2%; mean time to regression, 6.10 years). The rare cases of aggressive systemic mastocytosis were symptomatic from the outset. Congenital mastocytosis and the KIT D816V mutation were associated with CM regression (odds ratio, 0.48, P = .031, and 0.173, P = .031, respectively). Aggravation of MCASs over time was correlated with the persistence of skin lesions. However, the MCASs became more intense in 19% of the patients with MCASs at baseline and CM regression, justifying long-term follow-up in this setting. CONCLUSIONS Our results open up new hypotheses with regard to the spontaneous regression of CM in pediatric patients.
Collapse
|
26
|
Blank S, Grosch J, Ollert M, Bilò MB. Precision Medicine in Hymenoptera Venom Allergy: Diagnostics, Biomarkers, and Therapy of Different Endotypes and Phenotypes. Front Immunol 2020; 11:579409. [PMID: 33193378 PMCID: PMC7643016 DOI: 10.3389/fimmu.2020.579409] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Allergic reactions to stings of Hymenoptera species may be severe and are potentially fatal deviations of the immunological response observed in healthy individuals. However, venom-specific immunotherapy (VIT) is an immunomodulatory approach able to cure venom allergy in the majority of affected patients. An appropriate therapeutic intervention and the efficacy of VIT not only depend on a conclusive diagnosis, but might also be influenced by the patient-specific manifestation of the disease. As with other diseases, it should be borne in mind that there are different endotypes and phenotypes of venom allergy, each of which require a patient-tailored disease management and treatment scheme. Reviewed here are different endotypes of sting reactions such as IgE-mediated allergy, asymptomatic sensitization or a simultaneous presence of venom allergy and mast cell disorders including particular considerations for diagnosis and therapy. Additionally, phenotypical manifestations of venom allergy, as e.g. differences in age of onset and disease severity, multiple sensitization or patients unsusceptible to therapy, are described. Moreover, biomarkers and diagnostic strategies that might reflect the immunological status of the patient and their value for therapeutic guidance are discussed. Taken together, the increasing knowledge of different disease manifestations in venom hypersensitivity and the growing availability of diagnostic tools open new options for the classification of venom allergy and, hence, for personalized medical approaches and precision medicine in Hymenoptera venom allergy.
Collapse
Affiliation(s)
- Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Johannes Grosch
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Maria Beatrice Bilò
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy.,Allergy Unit, Department of Internal Medicine, University Hospital of Ancona, Ancona, Italy
| |
Collapse
|
27
|
Huang Y, Wei J, Huang X, Zhou W, Xu Y, Deng DH, Cheng P. Comprehensively analyze the expression and prognostic role for ten-eleven translocations (TETs) in acute myeloid leukemia. Transl Cancer Res 2020; 9:7259-7283. [PMID: 35117329 PMCID: PMC8798779 DOI: 10.21037/tcr-20-3149] [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: 09/29/2020] [Accepted: 11/06/2020] [Indexed: 12/02/2022]
Abstract
BACKGROUND The ten-eleven translocation (TET) family oxidize 5-methylcytosines (5mCs) and promote the locus-specific reversal of DNA. The role of TETs in acute myeloid leukemia (AML) is mostly unknown. METHODS TETs mRNA expression levels were analyzed via Gene Expression Profiling Interactive Analysis (GEPIA). The association TETs expression levels and methylation with prognosis by UALCAN GenomicScape, and METHsurv. We analyzed TETs' aberration types, located mutations, and structures via cBioPortal. GeneMANIA performed the functional network. Gene ontology (GO) enrichment was analyzed via LinkedOmics. MiWalK identified miRNAs, miTarbase, and TargetScan. Transcription factor (TF) targets were analyzed via ChEA3. GSCAlite analyzed the role of these defined genes in cancer pathways and potential drug targets. Finally, we selected AML patients in our department to investigate the mutated types of TETs. RESULTS TETs expression level results showed TET1 (P=0.003) and TET2 (P=0.004) overexpressed in Haferlach leukemia samples, TET3 (P=4.04e-8) downregulation in Andersson leukemia samples. TET2 and TET3 overexpression but TET1 downregulation in the GEPIA database. Overexpression of TET2 leads to positive outcomes (P=0.0091). The upregulation of TET2 led to poor survival for CN-AML patients, but downregulation of TET3 indicated a satisfactory prognosis. The hypermethylation of TETs like cg24705708 (P=0.036), cg05976228 (P=0.022), cg19127638 (P=0.022), cg15254238 (P=0.025), cg07669489 (P=0.037) indicate poor outcomes. Overexpression of GALNS (P=0.024) as an adverse biomarker, downregulation of E2F5 (P=0.037), MAP7 (P=0.019), and NRIP1 (P=0.0013) indicated good prognosis. Regulatory network analysis indicated TETs' functions, including covalent chromatin modification, histone modification, DNA methylation, or demethylation. Enrichment functions involving. TETs participate in several cancer pathways, including DNA repair response and receptor tyrosine kinase (RTK) signaling pathway. TETs are sensitive to belinostat, ceranib-2, docetaxel, tivantinib, and vincristine. CONCLUSION Present study showed that TETs have different expressions in AML, and the expression levels of TETs lead to different outcomes of AML. The TETs cancer pathway analysis will also provide potential therapy methods for AML patients with TETs aberrations.
Collapse
Affiliation(s)
- Yan Huang
- Department of Hematology and Rheumatology, People’s Hospital of Baise, Baise, China
| | - Jie Wei
- Department of Hematology and Rheumatology, People’s Hospital of Baise, Baise, China
| | - Xunjun Huang
- Department of Hematology and Rheumatology, People’s Hospital of Baise, Baise, China
| | - Weijie Zhou
- Department of Hematology and Rheumatology, People’s Hospital of Baise, Baise, China
| | - Yuling Xu
- Department of Hematology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dong-Hong Deng
- Department of Hematology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Peng Cheng
- Department of Hematology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
28
|
Serrao S, Firinu D, Olianas A, Deidda M, Contini C, Iavarone F, Sanna MT, Boroumand M, Amado F, Castagnola M, Messana I, Del Giacco S, Manconi B, Cabras T. Top-Down Proteomics of Human Saliva Discloses Significant Variations of the Protein Profile in Patients with Mastocytosis. J Proteome Res 2020. [PMID: 32575983 DOI: 10.1021/acs.jproteome.0c00207.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mastocytosis is a myeloproliferative neoplasm causing abnormal clonal mast cell accumulation in different tissues, such as skin and bone marrow. A cutaneous subtype (CM) is distinguished from a systemic one (SM); SM patients can be grouped into SM with (SM+C) or without (SM-C) additional cutaneous lesions, and their classification is often challenging. This study was purposed to highlight variations in the salivary proteome of patients with different mastocytosis subtypes and compared to healthy controls. A top-down proteomics approach coupled to a label-free quantitation revealed salivary profiles in patients different from those of controls and a down-regulation of peptides/proteins involved in the mouth homeostasis and defense, such as statherin, histatins, and acidic proline-rich proteins (aPRPs), and in innate immunity and inflammation, such as the cathepsin inhibitors, suggesting a systemic condition associated with an exacerbated inflammatory state. The up-regulation of antileukoproteinase and S100A8 suggested a protective role against the disease status. The two SM forms were distinguished by the lower levels of truncated forms of aPRPs, statherin, P-B peptide, and cystatin D and the higher levels of thymosin β4 and α-defensins 1 and 4 in SM-C patients with respect to SM+C. Data are available via ProteomeXchange with identifier PXD017759.
Collapse
Affiliation(s)
- Simone Serrao
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Davide Firinu
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09124 Cagliari, Italy
| | - Alessandra Olianas
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Margherita Deidda
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09124 Cagliari, Italy
| | - Cristina Contini
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Roma, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - M Teresa Sanna
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Mozhgan Boroumand
- Laboratorio di Proteomica e Metabonomica-IRCCS Fondazione Santa Lucia, 100168 Roma, Italy
| | - Francisco Amado
- QOPNA, Mass spectrometry center, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Massimo Castagnola
- Laboratorio di Proteomica e Metabonomica-IRCCS Fondazione Santa Lucia, 100168 Roma, Italy
| | - Irene Messana
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", Consiglio Nazionale delle Ricerche, 00185 Roma, Italy
| | - Stefano Del Giacco
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09124 Cagliari, Italy
| | - Barbara Manconi
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Tiziana Cabras
- Dipartimento di Scienze della Vita e dell'Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
29
|
Serrao S, Firinu D, Olianas A, Deidda M, Contini C, Iavarone F, Sanna MT, Boroumand M, Amado F, Castagnola M, Messana I, Del Giacco S, Manconi B, Cabras T. Top-Down Proteomics of Human Saliva Discloses Significant Variations of the Protein Profile in Patients with Mastocytosis. J Proteome Res 2020; 19:3238-3253. [PMID: 32575983 PMCID: PMC8008451 DOI: 10.1021/acs.jproteome.0c00207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Indexed: 01/06/2023]
Abstract
Mastocytosis is a myeloproliferative neoplasm causing abnormal clonal mast cell accumulation in different tissues, such as skin and bone marrow. A cutaneous subtype (CM) is distinguished from a systemic one (SM); SM patients can be grouped into SM with (SM+C) or without (SM-C) additional cutaneous lesions, and their classification is often challenging. This study was purposed to highlight variations in the salivary proteome of patients with different mastocytosis subtypes and compared to healthy controls. A top-down proteomics approach coupled to a label-free quantitation revealed salivary profiles in patients different from those of controls and a down-regulation of peptides/proteins involved in the mouth homeostasis and defense, such as statherin, histatins, and acidic proline-rich proteins (aPRPs), and in innate immunity and inflammation, such as the cathepsin inhibitors, suggesting a systemic condition associated with an exacerbated inflammatory state. The up-regulation of antileukoproteinase and S100A8 suggested a protective role against the disease status. The two SM forms were distinguished by the lower levels of truncated forms of aPRPs, statherin, P-B peptide, and cystatin D and the higher levels of thymosin β4 and α-defensins 1 and 4 in SM-C patients with respect to SM+C. Data are available via ProteomeXchange with identifier PXD017759.
Collapse
Affiliation(s)
- Simone Serrao
- Dipartimento
di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Davide Firinu
- Dipartimento
di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09124 Cagliari, Italy
| | - Alessandra Olianas
- Dipartimento
di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Margherita Deidda
- Dipartimento
di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09124 Cagliari, Italy
| | - Cristina Contini
- Dipartimento
di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Federica Iavarone
- Dipartimento
di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
- Fondazione
Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| | - M. Teresa Sanna
- Dipartimento
di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Mozhgan Boroumand
- Laboratorio
di Proteomica e Metabonomica-IRCCS Fondazione Santa Lucia, 100168 Roma, Italy
| | - Francisco Amado
- QOPNA, Mass
spectrometry center, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Massimo Castagnola
- Laboratorio
di Proteomica e Metabonomica-IRCCS Fondazione Santa Lucia, 100168 Roma, Italy
| | - Irene Messana
- Istituto
di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00185 Roma, Italy
| | - Stefano Del Giacco
- Dipartimento
di Scienze Mediche e Sanità Pubblica, Università di Cagliari, 09124 Cagliari, Italy
| | - Barbara Manconi
- Dipartimento
di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| | - Tiziana Cabras
- Dipartimento
di Scienze della Vita e dell’Ambiente, Università di Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
30
|
Betancor D, Plaza KL, Eguez JC, Nair P, Trus M. Exon 8 KIT mutation and pulmonary eosinophilia. Allergy 2020; 75:2094-2096. [PMID: 32168380 DOI: 10.1111/all.14272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Diana Betancor
- Division of Respirology McMaster University Hamilton ON Canada
| | | | - Jose Cano Eguez
- Division of Respirology McMaster University Hamilton ON Canada
| | - Parameswaran Nair
- Division of Respirology McMaster University Hamilton ON Canada
- Department of Medicine McMaster University Hamilton ON Canada
| | - Michael Trus
- Division of Hematology McMaster University Hamilton ON Canada
- Department of Pathology & Molecular Medicine McMaster University Hamilton ON Canada
| |
Collapse
|
31
|
Zhu L, Yang C, Zhong W, Huang YQ, Zhang Q, Xu WC, Chen YF. KIT-related piebaldism in a Chinese girl. Am J Med Genet A 2020; 182:1321-1328. [PMID: 32220041 DOI: 10.1002/ajmg.a.61576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/16/2020] [Accepted: 03/13/2020] [Indexed: 11/11/2022]
Abstract
Piebaldism is a rare, autosomal dominant and congenital pigmentary disorder characterized by stable depigmentation of the skin and white forelock. Mutations in KIT or SNAI2 genes result in piebaldism. Most individuals with piebaldism have a family history of the disorder. Herein, we report a 5-month-old Chinese girl with severe piebaldism but no family history thereof. She has white forelock and large patches of depigmentation in the jaw, central anterior trunk, perineum and extremities. We performed whole-exome and Sanger sequencing and identified a de novo KIT mutation (NM_000222.2: c.2657G>A, p.Gly886Val) in exon 18 of KIT in the proband. Currently, this mutation is located in the most extreme C-terminal of the tyrosine kinase domain 2 of the KIT gene amongst all reported mutations and causes a severe clinical phenotype. We further reviewed literature on piebaldism and summarized 79 KIT gene mutations that lead to this disease. Our study may expand knowledge on the genotype-phenotype correlation in piebaldism and serve as a reference for genetic counseling and prenatal diagnosis of affected families.
Collapse
Affiliation(s)
- Lei Zhu
- Guangdong Medical College, College of Dermatology, Anhui Medical University, Guangzhou, China.,Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Chao Yang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Wei Zhong
- Guangdong Medical College, College of Dermatology, Anhui Medical University, Guangzhou, China.,Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Yu-Qi Huang
- Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Qi Zhang
- Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Wen-Cong Xu
- Guangdong Medical College, College of Dermatology, Anhui Medical University, Guangzhou, China.,Guangdong Provincial Dermatology Hospital, Guangzhou, China
| | - Yong-Feng Chen
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Arase N, Wataya-Kaneda M, Murota H, Nakagawa Y, Yamaoka T, Itoi-Ochi S, Hirayasu K, Arase H, Fujimoto M, Katayama I. Genotype and phenotype analysis of patients with pediatric cutaneous mastocytosis, especially wild-type KIT patients. J Dermatol 2020; 47:426-429. [PMID: 32077120 DOI: 10.1111/1346-8138.15266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 01/20/2020] [Indexed: 11/26/2022]
Abstract
Pediatric cutaneous mastocytosis (CM) is mainly attributed to gain-of-function mutations in KIT in mast cells. On the other hand, growing evidence suggests that CM patients exist without KIT mutations. To date, the association between the KIT mutation status and clinical phenotype has not been elucidated in pediatric CM, especially in patients with wild-type KIT. Nevertheless, genetic analysis has yet to be performed with whole KIT sequence of mast cells in Japanese patients with pediatric CM. In the present study, 11 Japanese patients with pediatric CM were analyzed to determine whether they had KIT mutations in their skin, and their clinical phenotypes were observed. The approximate frequency of patients with KIT mutation and that of wild-type KIT was almost consistent with the European analysis. The distribution of overall macules was similar between the patients with and without KIT mutations.
Collapse
Affiliation(s)
- Noriko Arase
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroyuki Murota
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Dermatology, Nagasaki University Graduate School of Biomedical Science, Nagasaki, Japan
| | - Yukinobu Nakagawa
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshifumi Yamaoka
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan.,Nakatsu Dermatology Clinic, Osaka, Japan
| | - Saori Itoi-Ochi
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kouyuki Hirayasu
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Immunology, Graduate School of Medicine, Kanazawa University, Kanazawa, Japan.,WPI Nano Life Science Institute, Kanazawa University, Kanazawa, Japan
| | - Hisashi Arase
- Laboratory of Immunochemistry, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Immunochemistry, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan.,Laboratory of Cutaneous Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ichiro Katayama
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Pigmentation Research and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
33
|
Activation of Siglec-7 results in inhibition of in vitro and in vivo growth of human mast cell leukemia cells. Pharmacol Res 2020; 158:104682. [PMID: 32035162 DOI: 10.1016/j.phrs.2020.104682] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/31/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
Advanced systemic mastocytosis is a rare and still untreatable disease. Blocking antibodies against inhibitory receptors, also known as "immune checkpoints", have revolutionized anti-cancer treatment. Inhibitory receptors are expressed not only on normal immune cells, including mast cells but also on neoplastic cells. Whether activation of inhibitory receptors through monoclonal antibodies can lead to tumor growth inhibition remains mostly unknown. Here we show that the inhibitory receptor Siglec-7 is expressed by primary neoplastic mast cells in patients with systemic mastocytosis and by mast cell leukemia cell lines. Activation of Siglec-7 by anti-Siglec-7 monoclonal antibody caused phosphorylation of Src homology region 2 domain-containing phosphatase-1 (SHP-1), reduced phosphorylation of KIT and induced growth inhibition in mast cell lines. In SCID-beige mice injected with either the human mast cell line HMC-1.1 and HMC-1.2 or with Siglec-7 transduced B cell lymphoma cells, anti-Siglec-7 monoclonal antibody reduced tumor growth by a mechanism involving Siglec-7 cytoplasmic domains in "preventive" and "treatment" settings. These data demonstrate that activation of Siglec-7 on mast cell lines can inhibit their growth in vitro and in vivo. This might pave the way to additional treatment strategies for mastocytosis.
Collapse
|
34
|
Abstract
Mast cells and basophils play a crucial role during type I hypersensitivity reactions. However, despite efforts to elucidate their role in the pathogenesis of allergy and inflammation, our understanding of MC and basophil biology is still relatively scarce. The practical difficulty in obtaining a sufficient number of purified primary cells from biological samples has slowed down the process of reaching a full understanding of the physiological role of these functionally similar cell types. The establishment of several immortalized cell lines has been a useful tool to establish and perform sophisticated laboratory protocols that are impractical using primary cells. Continuous cell lines have been extensively used to investigate allergen/IgE-mediated cell activation, to elucidate the degranulation dynamics, to investigate structural and functional properties of the high-affinity receptor (FcεRI), and to test cell-stabilizing compounds. In this chapter, we review the most widely used and better-characterized MC and basophil cell lines, highlighting their advantages and drawbacks. It must be pointed out, however, that while cell lines represent a useful in vitro tool due to their easy manipulability and reduced culture costs, they often show aberrant characteristics which are not fully representative of primary cell physiology; results obtained with such cells therefore must be interpreted with due care.
Collapse
|
35
|
Rossignol J, Polivka L, Maouche-Chrétien L, Frenzel L, Dubreuil P, Hermine O. Recent advances in the understanding and therapeutic management of mastocytosis. F1000Res 2019; 8:F1000 Faculty Rev-1961. [PMID: 31824655 PMCID: PMC6880274 DOI: 10.12688/f1000research.19463.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
Mastocytosis is a rare disease due to the abnormal accumulation of mast cells in various tissues. Its clinical presentation is heterogeneous depending on mast cell infiltration and mediators release. In some cases, it is associated with hematological malignancies. Prognosis varies from very good with a life expectancy similar to the general population in indolent forms of the disease to a survival time of just a few months in mast cell leukemia. Although in most cases a somatic KIT D816V mutation is found in tumor mast cells, the physiopathology of the disease is not yet fully understood. Additional germline and somatic mutations may explain this heterogeneity. Treatments aim at blocking effect of mast cell mediators, reducing mast cell activation and tumor burden. New drugs mainly directed against the tyrosine kinase activity of KIT have dramatically changed the quality of life and prognosis of mast cell diseases. Present and future therapeutic strategies are discussed in this review.
Collapse
Affiliation(s)
- Julien Rossignol
- French Reference Center for Mastocytosis (CEREMAST), Necker Children's Hospital, APHP, Paris, France
| | - Laura Polivka
- Department of Hematology, Gustave Roussy Institute, Paris-Saclay University, Villejuif, France
| | | | - Laurent Frenzel
- Department of Dermatology, Necker Children's Hospital, APHP, Paris, France
| | - Patrice Dubreuil
- Department of Hematology, Necker Children's Hospital, APHP, Paris, France
| | - Olivier Hermine
- Centre de Recherche en Cancérologie de Marseille, Inserm U1068, Marseille, France
| |
Collapse
|
36
|
Impact of somatic and germline mutations on the outcome of systemic mastocytosis. Blood Adv 2019; 2:2814-2828. [PMID: 30373888 DOI: 10.1182/bloodadvances.2018020628] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 10/01/2018] [Indexed: 12/25/2022] Open
Abstract
Systemic mastocytosis (SM) is a highly heterogeneous disease with indolent and aggressive forms, with the mechanisms leading to malignant transformation still remaining to be elucidated. Here, we investigated the presence and frequency of genetic variants in 34 SM patients with multilineal KIT D816V mutations. Initial screening was performed by targeted sequencing of 410 genes in DNA extracted from purified bone marrow cells and hair from 12 patients with nonadvanced SM and 8 patients with advanced SM, followed by whole-genome sequencing (WGS) in 4 cases. Somatic mutations were further investigated in another 14 patients with advanced SM. Despite the fact that no common mutation other than KIT D816V was found in WGS analyses, targeted next-generation sequencing identified 67 nonsynonymous genetic variants involving 39 genes. Half of the mutations were somatic (mostly multilineal), whereas the other half were germline variants. The presence of ≥1 multilineal somatic mutation involving genes other than KIT D816V, ≥3 germline variants, and ≥1 multilineal mutation in the SRSF2, ASXL1, RUNX1, and/or EZH2 genes (S/A/R/E genes), in addition to skin lesions, splenomegaly, thrombocytopenia, low hemoglobin levels, and increased alkaline phosphatase and β2-microglobulin serum levels, were associated with a poorer patient outcome. However, the presence of ≥1 multilineal mutation, particularly involving S/A/R/E genes, was the only independent predictor for progression-free survival and overall survival in our cohort.
Collapse
|
37
|
Biasoli D, Compston-Garnett L, Ricketts SL, Birand Z, Courtay-Cahen C, Fineberg E, Arendt M, Boerkamp K, Melin M, Koltookian M, Murphy S, Rutteman G, Lindblad-Toh K, Starkey M. A synonymous germline variant in a gene encoding a cell adhesion molecule is associated with cutaneous mast cell tumour development in Labrador and Golden Retrievers. PLoS Genet 2019; 15:e1007967. [PMID: 30901340 PMCID: PMC6447235 DOI: 10.1371/journal.pgen.1007967] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 04/03/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023] Open
Abstract
Mast cell tumours are the most common type of skin cancer in dogs, representing a significant concern in canine health. The molecular pathogenesis is largely unknown, but breed-predisposition for mast cell tumour development suggests the involvement of inherited genetic risk factors in some breeds. In this study, we aimed to identify germline risk factors associated with the development of mast cell tumours in Labrador Retrievers, a breed with an elevated risk of mast cell tumour development. Using a methodological approach that combined a genome-wide association study, targeted next generation sequencing, and TaqMan genotyping, we identified a synonymous variant in the DSCAM gene on canine chromosome 31 that is associated with mast cell tumours in Labrador Retrievers. DSCAM encodes a cell-adhesion molecule. We showed that the variant has no effect on the DSCAM mRNA level but is associated with a significant reduction in the level of the DSCAM protein, suggesting that the variant affects the dynamics of DSCAM mRNA translation. Furthermore, we showed that the variant is also associated with mast cell tumours in Golden Retrievers, a breed that is closely related to Labrador Retrievers and that also has a predilection for mast cell tumour development. The variant is common in both Labradors and Golden Retrievers and consequently is likely to be a significant genetic contributor to the increased susceptibility of both breeds to develop mast cell tumours. The results presented here not only represent an important contribution to the understanding of mast cell tumour development in dogs, as they highlight the role of cell adhesion in mast cell tumour tumourigenesis, but they also emphasise the potential importance of the effects of synonymous variants in complex diseases such as cancer. The combination of various genetic and environmental risk factors makes the understanding of the molecular circuitry behind complex diseases, like cancer, a major challenge. The homogeneous nature of pedigree dog breed genomes makes these dogs ideal for the identification of both simple disease-causing genetic variants and genetic risk factors for complex diseases. Mast cell tumours are the most common type of canine skin cancer, and one of the most common cancers affecting dogs of most breeds. Several breeds, including Labrador Retrievers (which represent one of the most popular dog breeds), have an elevated risk of mast cell tumour development. Here, by using a methodological approach that combined different techniques, we identified a common inherited synonymous variant, that predisposes Labrador Retrievers to mast cell tumour development. Interestingly, we showed that this variant, despite its synonymous nature, appears to have an effect on translation dynamics as it is associated with reduced levels of DSCAM, a cell adhesion molecule. The results presented here reveal dysregulation of cell adhesion to be an important factor in mast cell tumour pathogenesis, and also highlight the important role that synonymous variants can play in complex diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Maja Arendt
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Kim Boerkamp
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
| | - Malin Melin
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Michele Koltookian
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Sue Murphy
- Animal Health Trust, Newmarket, United Kingdom
| | - Gerard Rutteman
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, The Netherlands
- Veterinary Specialist Centre De Wagenrenk, Wageningen, The Netherlands
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
| | - Mike Starkey
- Animal Health Trust, Newmarket, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
|
39
|
Abstract
Mast cell disorders comprise a heterogeneous group of rare diseases, the diagnosis of which still remains a challenge. Bone marrow analysis constitutes the most appropriate site for screening systemic involvement in mastocytosis. Morphologic, immunohistochemical, flow cytometric immunophenotyping, and molecular studies should be routinely performed for diagnostic/prognostic purposes in experienced reference centers during the diagnostic workup in suspected systemic mastocytosis. The authors review the most relevant characteristics of bone marrow expression of mast cell disorders as well as the different methodological approaches to be applied to perform an objective and reproducible diagnosis and classification of mastocytosis and other mast cell disorders.
Collapse
|
40
|
Kholodenko IV, Kalinovsky DV, Doronin II, Deyev SM, Kholodenko RV. Neuroblastoma Origin and Therapeutic Targets for Immunotherapy. J Immunol Res 2018; 2018:7394268. [PMID: 30116755 PMCID: PMC6079467 DOI: 10.1155/2018/7394268] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/27/2018] [Indexed: 01/30/2023] Open
Abstract
Neuroblastoma is a pediatric solid cancer of heterogeneous clinical behavior. The unique features of this type of cancer frequently hamper the process of determining clinical presentation and predicting therapy effectiveness. The tumor can spontaneously regress without treatment or actively develop and give rise to metastases despite aggressive multimodal therapy. In recent years, immunotherapy has become one of the most promising approaches to the treatment of neuroblastoma. Still, only one drug for targeted immunotherapy of neuroblastoma, chimeric monoclonal GD2-specific antibodies, is used in the clinic today, and its application has significant limitations. In this regard, the development of effective and safe GD2-targeted immunotherapies and analysis of other potential molecular targets for the treatment of neuroblastoma represents an important and topical task. The review summarizes biological characteristics of the origin and development of neuroblastoma and outlines molecular markers of neuroblastoma and modern immunotherapy approaches directed towards these markers.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow 119121, Russia
| | - Daniel V. Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Igor I. Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Real Target LLC, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Sergey M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University “MEPhI”, Moscow 115409, Russia
| | - Roman V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Real Target LLC, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| |
Collapse
|
41
|
Dahlin JS, Hamey FK, Pijuan-Sala B, Shepherd M, Lau WWY, Nestorowa S, Weinreb C, Wolock S, Hannah R, Diamanti E, Kent DG, Göttgens B, Wilson NK. A single-cell hematopoietic landscape resolves 8 lineage trajectories and defects in Kit mutant mice. Blood 2018; 131:e1-e11. [PMID: 29588278 PMCID: PMC5969381 DOI: 10.1182/blood-2017-12-821413] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/16/2018] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) maintain the adult blood system, and their dysregulation causes a multitude of diseases. However, the differentiation journeys toward specific hematopoietic lineages remain ill defined, and system-wide disease interpretation remains challenging. Here, we have profiled 44 802 mouse bone marrow HSPCs using single-cell RNA sequencing to provide a comprehensive transcriptional landscape with entry points to 8 different blood lineages (lymphoid, megakaryocyte, erythroid, neutrophil, monocyte, eosinophil, mast cell, and basophil progenitors). We identified a common basophil/mast cell bone marrow progenitor and characterized its molecular profile at the single-cell level. Transcriptional profiling of 13 815 HSPCs from the c-Kit mutant (W41/W41) mouse model revealed the absence of a distinct mast cell lineage entry point, together with global shifts in cell type abundance. Proliferative defects were accompanied by reduced Myc expression. Potential compensatory processes included upregulation of the integrated stress response pathway and downregulation of proapoptotic gene expression in erythroid progenitors, thus providing a template of how large-scale single-cell transcriptomic studies can bridge between molecular phenotypes and quantitative population changes.
Collapse
Affiliation(s)
- Joakim S Dahlin
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Fiona K Hamey
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Blanca Pijuan-Sala
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Mairi Shepherd
- Department of Haematology, University of Cambridge, Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; and
| | - Winnie W Y Lau
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Sonia Nestorowa
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Caleb Weinreb
- Department of Systems Biology, Harvard Medical School, Boston, MA
| | - Samuel Wolock
- Department of Systems Biology, Harvard Medical School, Boston, MA
| | - Rebecca Hannah
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Evangelia Diamanti
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - David G Kent
- Department of Haematology, University of Cambridge, Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom; and
| | - Berthold Göttgens
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Nicola K Wilson
- Department of Haematology, University of Cambridge, Cambridge Institute for Medical Research and Wellcome-Medical Research Council Cambridge Stem Cell Institute, Cambridge, United Kingdom
| |
Collapse
|
42
|
Chan IJ, Tharp MD. Comparison of lesional skin c-KIT mutations with clinical phenotype in patients with mastocytosis. Clin Exp Dermatol 2018; 43:416-422. [PMID: 29350409 DOI: 10.1111/ced.13362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Activating c-KIT mutations cause abnormal mast cell growth and appear to play a role in mastocytosis. However, the correlation of c-KIT mutations with disease phenotypes is poorly characterized. AIM To evaluate the correlation of c-KIT mutations with clinical presentations and laboratory findings. METHODS Total cellular RNA was isolated from the skin lesions of 43 adults and 7 children with mastocytosis, and PCR amplicons of cDNA were sequenced for c-KIT mutations. RESULTS The most common activating mutation, KIT-D816V, was identified in 72% of adults and 57% of children. Additional activating mutations, namely, V560G and the internal tandem duplications (ITDs) 502-503dupAY, were detected in 12% of adults and 8% of children. V560G occurred more commonly in our patients than previously reported, and it appeared to be associated with more advanced disease. Otherwise, the presence or absence of activating mutations did not correlate with skin lesion morphology, disease extent or total serum tryptase levels. Four adults had expression only of wild-type KIT, while two others had expression of a truncated KIT lacking tyrosine kinase activity; yet these patients were clinically indistinguishable from those patients with activating c-KIT mutations. CONCLUSIONS Activating c-KIT mutations exist in a significant portion of patients with mastocytosis, but not all patients showed expression of these mutations. Except for V560G, the presence or absence of activating c-KIT mutations did not predict the extent of disease. These observations suggest that although activating c-KIT mutations are associated with mast cell growth, other genes probably play a role in the cause of mastocytosis.
Collapse
Affiliation(s)
- I J Chan
- Department of Dermatology, Rush University Medical Center, Chicago, IL, USA
| | - M D Tharp
- Department of Dermatology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
43
|
Greiner G, Gurbisz M, Ratzinger F, Witzeneder N, Simonitsch-Klupp I, Mitterbauer-Hohendanner G, Mayerhofer M, Müllauer L, Sperr WR, Valent P, Hoermann G. Digital PCR: A Sensitive and Precise Method for KIT D816V Quantification in Mastocytosis. Clin Chem 2017; 64:547-555. [PMID: 29237714 DOI: 10.1373/clinchem.2017.277897] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 11/28/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND The analytically sensitive detection of KIT D816V in blood and bone marrow is important for diagnosing systemic mastocytosis (SM). Additionally, precise quantification of the KIT D816V variant allele fraction (VAF) is relevant clinically because it helps to predict multilineage involvement and prognosis in cases of advanced SM. Digital PCR (dPCR) is a promising new method for sensitive detection and accurate quantification of somatic mutations. METHODS We performed a validation study of dPCR for KIT D816V on 302 peripheral blood and bone marrow samples from 156 patients with mastocytosis for comparison with melting curve analysis after peptide nucleic acid-mediated PCR clamping (clamp-PCR) and allele-specific quantitative real-time PCR (qPCR). RESULTS dPCR showed a limit of detection of 0.01% VAF with a mean CV of 8.5% and identified the mutation in 90% of patients compared with 70% for clamp-PCR (P < 0.001). Moreover, dPCR for KIT D816V was highly concordant with qPCR without systematic deviation of results, and confirmed the clinical value of KIT D816V VAF measurements. Thus, patients with advanced SM showed a significantly higher KIT D816V VAF (median, 2.43%) compared with patients with indolent SM (median, 0.14%; P < 0.001). Moreover, dPCR confirmed the prognostic significance of a high KIT D816V VAF regarding survival (P < 0.001). CONCLUSIONS dPCR for KIT D816V provides a high degree of precision and sensitivity combined with the potential for interlaboratory standardization, which is crucial for the implementation of KIT D816V allele burden measurement. Thus, dPCR is suitable as a new method for KIT D816V testing in patients with mastocytosis.
Collapse
Affiliation(s)
- Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Gurbisz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Franz Ratzinger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nadine Witzeneder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.,Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | | | | | - Leonhard Müllauer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; .,Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW In this review, we examine the current understanding of the pathogenesis, clinical presentations, diagnostic tools, and treatment options of pediatric mastocytosis as well as the natural history of the disease. RECENT FINDINGS We discuss the emerging concept of mast cell activation syndrome. Mastocytosis in children presents most commonly as isolated cutaneous lesions and is a relatively rare occurrence with excellent prognosis and spontaneous regression often occurring by adolescence. Systemic mastocytosis with organ system involvement is a more serious condition and is likely to persist into adulthood.
Collapse
Affiliation(s)
- Nicholas Klaiber
- Department of Pediatrics, Division of Allergy and Immunology, Virginia Commonwealth University Health Systems, 1000 E Broad Street, Richmond, VA, 23219, USA
| | - Santhosh Kumar
- Department of Pediatrics, Division of Allergy and Immunology, Virginia Commonwealth University Health Systems, 1000 E Broad Street, Richmond, VA, 23219, USA
| | - Anne-Marie Irani
- Department of Pediatrics, Division of Allergy and Immunology, Virginia Commonwealth University Health Systems, 1000 E Broad Street, Richmond, VA, 23219, USA.
| |
Collapse
|
45
|
Abstract
The classification "gastrointestinal stromal tumor" (GIST) became commonplace in the 1990s and since that time various advances have characterized the GIST lineage of origin, tyrosine kinase mutations, and mechanisms of response and resistance to targeted therapies. In addition to tyrosine kinase mutations and their constitutive activation of downstream signaling pathways, GISTs acquire a sequence of chromosomal aberrations. These include deletions of chromosomes 14q, 22q, 1p, and 15q, which harbor putative tumor suppressor genes required for stepwise progression from microscopic, preclinical forms of GIST (microGIST) to clinically relevant tumors with malignant potential. Recent advances extend our understanding of GIST biology beyond that of the oncogenic KIT/PDGFRA tyrosine kinases and beyond mechanisms of KIT/PDGFRA-inhibitor treatment response and resistance. These advances have characterized ETV1 as an essential interstitial cell of Cajal-GIST transcription factor in oncogenic KIT signaling pathways, and have characterized the biologically distinct subgroup of succinate dehydrogenase deficient GIST, which are particularly common in young adults. Also, recent discoveries of MAX and dystrophin genomic inactivation have expanded our understanding of GIST development and progression, showing that MAX inactivation is an early event fostering cell cycle activity, whereas dystrophin inactivation promotes invasion and metastasis.
Collapse
|
46
|
Metcalfe DD, Mekori YA. Pathogenesis and Pathology of Mastocytosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:487-514. [PMID: 28135563 DOI: 10.1146/annurev-pathol-052016-100312] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Systemic mastocytosis is a clonal disorder of mast cells that may variably present with characteristic skin lesions, episodes of mast cell mediator release, and disturbances of hematopoiesis. No curative therapy presently exists. Conventional management has relied on agents that antagonize mediators released by mast cells, inhibit mediator secretion, or modulate mast cell proliferation. Recent advances in the molecular understanding of the pathophysiology of systemic mastocytosis have provided new therapeutic considerations, including new and novel tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Dean D Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892;
| | - Yoseph A Mekori
- Tel Hai College, Upper Galilee, 1220800 Israel; .,Meir Medical Center, Kfar Saba 44281, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
47
|
Vaes M, Benghiat FS, Hermine O. Targeted Treatment Options in Mastocytosis. Front Med (Lausanne) 2017; 4:110. [PMID: 28775983 PMCID: PMC5517467 DOI: 10.3389/fmed.2017.00110] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022] Open
Abstract
Mastocytosis refers to a heterogeneous group of disorders resulting from the clonal proliferation of abnormal mast cells and their accumulation in the skin (cutaneous mastocytosis when only in the skin, CM) or in various organs (systemic mastocytosis, SM). This leads to a wide variety of clinical manifestations resulting from excessive mediator release in CM and benign forms of SM (indolent SM, ISM) and from tissue mast cell infiltration causing multiorgan dysfunction and failure in more aggressive subtypes (aggressive SM, ASM, or mast cell leukemia). In addition, SM may be associated with hematological neoplasms (AHN). While treatment of ISM primarily aims at symptom management with anti-mediator therapies, cytoreductive and targeted therapies are needed to control the expansion of neoplastic mast cells in advanced forms of SM, in order to improve overall survival. Mast cell accumulation results from a gain-of-function mutation (mostly the D816V mutation) within the KIT tyrosine kinase domain expressed by mast cells and additional genetic and epigenetic mutations may further determine the features of the disease (ASM and AHN). Consequently, tyrosine kinase inhibitors and targeted therapies directed against the oncogenic signaling machinery downstream of KIT are attractive therapeutic approaches. A better understanding of the relative contribution of these genetic and epigenetic events to the molecular pathogenesis of mastocytosis is of particular interest for the development of targeted therapies and therefore to better choose patient subgroups that would best benefit from a given therapeutic strategy.
Collapse
Affiliation(s)
- Mélanie Vaes
- Department of Hematology, Université Libre de Bruxelles, Hopital Erasme, Brussels, Belgium.,Department of Hematology, Université Libre de Bruxelles, CHU Tivoli, La Louvière, Belgium
| | | | - Olivier Hermine
- French Reference Center for Mastocytosis (CEREMAST), Department of Hematology, Necker Children's Hospital, APHP, Paris, France.,Imagine Institute for Genetic Diseases (INSERM U1163 CNRS ERL 8654), Paris Descartes University, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
48
|
Astle JM, Rose MG, Racke FK, Tormey CA, Siddon AJ. R634W KIT Mutation in an Adult With Systemic Mastocytosis. Lab Med 2017; 48:253-257. [DOI: 10.1093/labmed/lmx026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Indexed: 12/25/2022] Open
|
49
|
Sellar R, Losman JA. Targeting Aberrant Signaling in Myeloid Malignancies: Promise Versus Reality. Hematol Oncol Clin North Am 2017; 31:565-576. [PMID: 28673388 DOI: 10.1016/j.hoc.2017.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Clonal myeloid disorders are characterized by genetic alterations that activate cytokine signaling pathways and stimulate cell proliferation. These activated signaling pathways have been extensively studied as potential therapeutic targets, and tyrosine kinase inhibitors have indeed had extraordinary success in treating BCR/ABL-positive chronic myeloiud leukemia. However, although inhibitors of other activated kinases have been developed that perform well in preclinical studies, the therapeutic efficacy of these drugs in patients has been unimpressive. This article discusses potential reasons for these discordant results and outlines recent scientific advances that are informing future efforts to target activated kinases in clonal myeloid disorders.
Collapse
Affiliation(s)
- Rob Sellar
- Division of Hematology, Brigham and Women's Hospital, 1 Blackfan Circle, Karp Building, CHRB05.125, Boston, MA 02115, USA
| | - Julie-Aurore Losman
- Department of Medical Oncology, Division of Hematology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
50
|
Malik F, Ali N, Jafri SIM, Ghani A, Hamid M, Boigon M, Fidler C. Continuous diphenhydramine infusion and imatinib for KIT-D816V-negative mast cell activation syndrome: a case report. J Med Case Rep 2017; 11:119. [PMID: 28438191 PMCID: PMC5402659 DOI: 10.1186/s13256-017-1278-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/25/2017] [Indexed: 12/01/2022] Open
Abstract
Background We present the first full case report of the treatment of mast cell activation syndrome with continuous diphenhydramine infusion, which resulted in the improvement of anaphylactic reactions and a decrease in hospital readmission. Furthermore, the patient received imatinib in the absence of the KIT-D816V mutation, which led to further improvement of quality of life. Currently, we are trying to wean this patient off diphenhydramine; if successful, this attempt will represent the first reported case. Case presentation An 18-year-old white girl presented with a flare of mast cell activation syndrome and received epinephrine and steroids. She had failed multiple previous therapies, and her quality of life was affected due to two to three flares/week. She was started on continuous diphenhydramine infusion and imatinib, which led to a decrease in hospital admissions and marked improvement in her quality of life. Conclusions Continuous diphenhydramine infusion can provide promising outcomes following the failure of intermittent antihistamine dosing in patients with severe mast cell activation syndrome. Initiating continuous diphenhydramine infusion may be helpful in an intensive care setting when the patient is particularly prone to anaphylaxis and/or the resources needed to manage anaphylaxis are not available outside the intensive care unit. Furthermore, imatinib provides benefits in KIT-D816V-negative mast cell disorders due to other unknown mutations.
Collapse
Affiliation(s)
- Faizan Malik
- Department of Internal Medicine, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA.
| | - Naveed Ali
- Department of Internal Medicine, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA
| | - Syed Imran Mustafa Jafri
- Department of Internal Medicine, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA
| | - Ali Ghani
- Department of Internal Medicine, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA
| | - Mohsin Hamid
- Department of Internal Medicine, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA
| | - Margot Boigon
- Department of Internal Medicine, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA
| | - Christian Fidler
- Rosenfeld Cancer Center, Abington Jefferson Health, 1200 Old York Road, Abington, PA, 19001, USA
| |
Collapse
|