1
|
Stewart BL, Helber H, Bannon SA, Deuitch NT, Ferguson M, Fiala E, Hamilton KV, Malcolmson J, Pencheva B, Smith-Simmer K. Risk assessment and genetic counseling for hematologic malignancies-Practice resource of the National Society of Genetic Counselors. J Genet Couns 2025; 34:e1959. [PMID: 39189353 DOI: 10.1002/jgc4.1959] [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: 11/22/2023] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024]
Abstract
Hematologic malignancies (HMs) are a heterogeneous group of cancers impacting individuals of all ages that have been increasingly recognized in association with various germline predisposition syndromes. Given the myriad of malignancy subtypes, expanding differential diagnoses, and unique sample selection requirements, evaluation for hereditary predisposition to HM presents both challenges as well as exciting opportunities in the ever-evolving field of genetic counseling. This practice resource has been developed as a foundational resource for genetic counseling approaches to hereditary HMs and aims to empower genetic counselors who encounter individuals and families with HMs in their practice.
Collapse
Affiliation(s)
| | - Hannah Helber
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Hematology and Cancer Center, Texas Children's Hospital, Houston, Texas, USA
| | - Sarah A Bannon
- National Institute of Allergy & Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Natalie T Deuitch
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Elise Fiala
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kayla V Hamilton
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| | - Janet Malcolmson
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bojana Pencheva
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Kelcy Smith-Simmer
- Academic Affairs, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, UW Health, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Abramowicz S, Chabbi I, Fardeau C, Touhami S, Bodaghi B. CBL syndrome presenting with severe EBV infection and panuveitis masquerade. Eur J Ophthalmol 2025; 35:NP1-NP7. [PMID: 39604255 DOI: 10.1177/11206721241302113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
PURPOSE To describe a case of CBL syndrome presenting with life-threatening pediatric Epstein-Barr virus (EBV) infection and sight-threatening rod-cone dystrophy (RCD) masquerading as severe panuveitis. METHODS Single case report with results of whole-exome sequencing (WES) analysis in the proband and his parents. Data were collected from July 2009 to December 2022. RESULTS A 35-month-old boy presented with severe primary EBV infection complicated by encephalitis and hemophagocytic lymphohistiocytosis. The clinical picture abated with systemic corticosteroids. Genetic testing revealed a heterozygous germline pathogenic variant in the CBL gene c.1141T > G (p.Cys381Gly). At 6 years old, he developed a severe bilateral panuveitis requiring multiple lines of immunosuppressants, mostly to control refractory cystoid macular edema (CME). During follow-up, intraretinal pigment deposits and peripheral retinal atrophy started to appear. Full-field electroretinogram (ffERG) revealed a pattern consistent with RCD. Repeat WES targeting known inherited retinal disease (IRD) genes was negative. A diagnosis of CBL syndrome complicated by RCD masquerading as severe bilateral panuveitis was made, and CME treatment was switched to oral acetazolamide. CONCLUSIONS CBL syndrome can present with severe EBV infection early in life. RCD masquerading as severe panuveitis is also a possible feature of CBL syndrome. RCD should be kept in mind in patients with this syndrome who present with idiopathic intraocular inflammation and are refractory to IMT. Carbonic anhydrase inhibitors (CAI) should be tried early to treat CME.
Collapse
Affiliation(s)
- Stéphane Abramowicz
- Department of Ophthalmology, IHU FOReSIGHT, Hôpital Universitaire Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Inès Chabbi
- Department of Ophthalmology, IHU FOReSIGHT, Hôpital Universitaire Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Christine Fardeau
- Department of Ophthalmology, IHU FOReSIGHT, Hôpital Universitaire Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Sara Touhami
- Department of Ophthalmology, IHU FOReSIGHT, Hôpital Universitaire Pitié-Salpêtrière, Sorbonne Université, Paris, France
| | - Bahram Bodaghi
- Department of Ophthalmology, IHU FOReSIGHT, Hôpital Universitaire Pitié-Salpêtrière, Sorbonne Université, Paris, France
| |
Collapse
|
3
|
Currao P, Balzarini M, Pruna D, Marica M, Soddu C, Marras M, Pavanello M, Satta S, Savasta S. Vascular Abnormalities and Neurofibromatosis Type 1: A Paediatric Case Series. J Child Neurol 2025; 40:49-60. [PMID: 39380389 DOI: 10.1177/08830738241284081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Neurofibromatosis type 1 (NF1) is a multisystemic neurocutaneous disease caused by a heterozygous mutation of the NF1 gene that encodes neurofibromin. Complications include vascular and neurologic abnormalities such as moyamoya syndrome, a cerebrovascular disorder with progressive occlusion of the large intracranial arteries, leading to ischemic events and the formation of abnormal vascular networks. Stenosis of the renal artery is another frequent complication of neurofibromatosis type 1, and it represents the most common cause of secondary hypertension in these patients. The purpose of the article is to describe the clinical manifestations of neurofibromatosis type 1 vasculopathy in 4 patients presenting with a wide range of neurologic and reno-vascular manifestations, as well as to examine current diagnostic management and follow-up, current therapeutic options, and to discuss further perspectives in terms of screening, diagnosis, and treatment.
Collapse
Affiliation(s)
- Paolo Currao
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| | - Marta Balzarini
- Pediatric Service, San Michele Hospital, ARNAS Brotzu Cagliari, Cagliari, Italy
| | - Dario Pruna
- Department of Pediatric Neurology, "Microcitemico - A. Cao" Pediatric Hospital, ASL Cagliari, Cagliari, Italy
| | - Monica Marica
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| | - Consolata Soddu
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| | - Mariangela Marras
- Department of Pediatric Radiology, "Microcitemico - A. Cao" Hospital, ASL Cagliari, Cagliari, Italy
| | - Marco Pavanello
- Department of Neurosurgery, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini, Genoa, Italy
| | - Stefania Satta
- Department of Medical Science and Public Health, University of Cagliari, Laboratory of Genetics and Genomics, "Microcitemico - A. Cao" Pediatric Hospital, ASL Cagliari, Cagliari, Italy
| | - Salvatore Savasta
- Pediatric Clinic, "Microcitemico - A. Cao" Pediatric Hospital, University of Cagliari, Cagliari, Italy
| |
Collapse
|
4
|
Arrabito M, Li Volsi N, La Rosa M, Samperi P, Pulvirenti G, Cannata E, Russo G, Di Cataldo A, Lo Nigro L. Transient Myeloproliferative Disorder (TMD), Acute Lymphoblastic Leukemia (ALL), and Juvenile Myelomonocytic Leukemia (JMML) in a Child with Noonan Syndrome: Sequential Occurrence, Single Center Experience, and Review of the Literature. Genes (Basel) 2024; 15:1191. [PMID: 39336782 PMCID: PMC11431238 DOI: 10.3390/genes15091191] [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: 08/16/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Noonan syndrome (NS) is an autosomal dominant disorder that varies in severity and can involve multiple organ systems. In approximately 50% of cases, it is caused by missense mutations in the PTPN11 gene (12q24.13). NS is associated with a higher risk of cancer occurrence, specifically hematological disorders. Here, we report a case of a child who was diagnosed at birth with a transient myeloproliferative disorder (TMD). After two years, the child developed hyperdiploid B-cell precursor acute lymphoblastic leukemia (BCP-ALL), receiving a two-year course of treatment. During her continuous complete remission (CCR), a heterozygous germline mutation in the PTPN11 gene [c.218 C>T (p.Thr73lle)] was identified. At the age of ten, the child presented with massive splenomegaly, hyperleukocytosis, and thrombocytopenia, resulting in the diagnosis of juvenile myelomonocytic leukemia (JMML). After an initial response to antimetabolite therapy (6-mercaptopurine), she underwent haploidentical hematopoietic stem cell transplantation (HSCT) and is currently in complete remission. The goal of this review is to gain insight into the various hematological diseases associated with NS, starting from our unique case.
Collapse
Affiliation(s)
- Marta Arrabito
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy
| | - Nicolò Li Volsi
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
- School of Medical Genetics, University of Catania, 95100 Catania, Italy
| | - Manuela La Rosa
- Cytogenetic-Cytofluorimetric-Molecular Biology Lab, Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, Via Santa Sofia 78, 95123 Catania, Italy;
| | - Piera Samperi
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
| | | | - Emanuela Cannata
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy
| | - Giovanna Russo
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy
- School of Pediatrics, University of Catania, 95100 Catania, Italy;
| | - Andrea Di Cataldo
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
- Department of Clinical and Experimental Medicine, University of Catania, 95100 Catania, Italy
- School of Pediatrics, University of Catania, 95100 Catania, Italy;
| | - Luca Lo Nigro
- Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, 95100 Catania, Italy; (M.A.); (N.L.V.); (P.S.); (E.C.); (G.R.); (A.D.C.)
- Cytogenetic-Cytofluorimetric-Molecular Biology Lab, Center of Pediatric Hematology Oncology, Azienda Policlinico di Catania, Via Santa Sofia 78, 95123 Catania, Italy;
| |
Collapse
|
5
|
Zong H, Huang Y, Xiong Y, Gong W, Lin B, Yang C. Severe generalized edema in a premature neonate: A case report and literature review. Clin Case Rep 2024; 12:e9341. [PMID: 39229301 PMCID: PMC11369640 DOI: 10.1002/ccr3.9341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 09/05/2024] Open
Abstract
Key Clinical Message With no family history, and an atypical phenotype, the clinical diagnosing of Noonan syndrome (NS) can be very difficult. The present case emphasized that generalized edema in neonates may be the potential first symptom of NS. Abstract Severe generalized edema is a rare pathological condition with high mortality in newborns, in particular the premature infants. It is characterized by the extensive subcutaneous tissue edema and the accumulation of fluid in neonatal body fluid compartments. The etiology and pathogenesis of hydrops in neonates are quite complex. Generally speaking, hydrops can be divided into immune hydrops and non-immune hydrops according to the etiology. It is still challenging in treating severe neonatal edema. In this study, we presented a preterm newborn with severe generalized edema after birth, which was finally diagnosed with Noonan syndrome (NS). The infant clinically manifested as severe generalized edema alone, without the involvement of multiple organ malformation. Generalized edema in neonates was probably the first symptom of NS. Therefore, differential diagnosis of NS is necessary for infants developing generalized edema.
Collapse
Affiliation(s)
- Haifeng Zong
- Neonatal Intensive Care UnitShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Yingsui Huang
- Neonatal Intensive Care UnitShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Ying Xiong
- Neonatal Intensive Care UnitShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Wentao Gong
- Neonatal Intensive Care UnitShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Bingchun Lin
- Neonatal Intensive Care UnitShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Chuanzhong Yang
- Neonatal Intensive Care UnitShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| |
Collapse
|
6
|
Niemeyer CM, Erlacher M. SH2B3 alterations in a novel genetic condition, juvenile myelomonocytic leukemia, and myeloproliferative neoplasia. Haematologica 2024; 109:2391-2394. [PMID: 38618667 PMCID: PMC11290542 DOI: 10.3324/haematol.2023.284747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg.
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg
| |
Collapse
|
7
|
Arfeuille C, Vial Y, Cadenet M, Caye-Eude A, Fenneteau O, Neven Q, Bonnard AA, Pizzi S, Carpentieri G, Capri Y, Girardi K, Pedace L, Macchiaiolo M, Boudhar K, Khaled MB, Chahla WA, Lutun A, Fahd M, Drunat S, Flex E, Dalle JH, Strullu M, Locatelli F, Tartaglia M, Cavé H. Germline bi-allelic SH2B3/LNK alteration predisposes to a neonatal juvenile myelomonocytic leukemia-like disorder. Haematologica 2024; 109:2542-2554. [PMID: 37981895 PMCID: PMC11290538 DOI: 10.3324/haematol.2023.283917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare, generally aggressive myeloproliferative neoplasm affecting young children. It is characterized by granulomonocytic expansion, with monocytosis infiltrating peripheral tissues. JMML is initiated by mutations upregulating RAS signaling. Approximately 10% of cases remain without an identified driver event. Exome sequencing of two unrelated cases of familial JMML of unknown genetics and analysis of the French JMML cohort identified 11 patients with variants in SH2B3, encoding LNK, a negative regulator of the JAK-STAT pathway. All variants were absent from healthy population databases, and the mutation spectrum was consistent with a loss of function of the LNK protein. A stoploss variant was shown to affect both protein synthesis and stability. The other variants were either truncating or missense, the latter affecting the SH2 domain that interacts with activated JAK. Of the 11 patients, eight from five families inherited pathogenic bi-allelic SH2B3 germline variants from their unaffected heterozygous parents. These children represent half of the cases with no identified causal mutation in the French cohort. They displayed typical clinical and hematologic features of JMML with neonatal onset and marked thrombocytopenia. They had a hypomethylated DNA profile with fetal characteristics and did not have additional genetic alterations. All patients showed partial or complete spontaneous clinical resolution. However, progression to thrombocythemia and immunity-related pathologies may be of concern later in life. Bi-allelic SH2B3 germline mutations thus define a new condition predisposing to a JMML-like disorder, suggesting that JAK pathway deregulation is capable of initiating JMML, and opening new therapeutic options.
Collapse
Affiliation(s)
- Chloé Arfeuille
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Yoann Vial
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Margaux Cadenet
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Aurélie Caye-Eude
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Odile Fenneteau
- Service d'Hématologie Biologique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Quentin Neven
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Adeline A Bonnard
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris
| | - Simone Pizzi
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Giovanna Carpentieri
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Yline Capri
- Département de Génétique, Unité de Génétique clinique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Katia Girardi
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Lucia Pedace
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Marina Macchiaiolo
- Rare Diseases and Medical Genetics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Kamel Boudhar
- Service de réanimation néonatale, Hôpital Central de l'Armée, Alger, Algérie
| | - Monia Ben Khaled
- University of Tunis El Manar, Faculty of Medicine of Tunis, 1007, Tunisia. Pediatric Immuno- Hematology Unit, Bone Marrow Transplantation Center Tunis, Tunis, Tunisia
| | - Wadih Abou Chahla
- Service d'Hématologie Pédiatrique, Centre Hospitalier Universitaire de Lille, Lille
| | - Anne Lutun
- Service d'Hématologie Pédiatrique, Centre Hospitalier Universitaire d'Amiens, Amiens
| | - Mony Fahd
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Séverine Drunat
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome
| | - Jean-Hugues Dalle
- Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Marion Strullu
- INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France; Service d'Onco-Hématologie pédiatrique, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy; Department of Pediatrics, Catholic University of the Sacred Hearth, 00168 Rome
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Bambino Gesù Children's Hospital IRCCS, 00146 Rome
| | - Hélène Cavé
- Département de Génétique, Unité de Génétique Moléculaire, Hôpital Robert Debré, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; INSERM UMR_S1131, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris.
| |
Collapse
|
8
|
Fuentes-Mateos R, García-Navas R, Fernández-Infante C, Hernández-Cano L, Calzada-Nieto N, Juan AOS, Guerrero C, Santos E, Fernández-Medarde A. Combined HRAS and NRAS ablation induces a RASopathy phenotype in mice. Cell Commun Signal 2024; 22:332. [PMID: 38886790 PMCID: PMC11184836 DOI: 10.1186/s12964-024-01717-4] [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: 02/05/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND HRASKO/NRASKO double knockout mice exhibit exceedingly high rates of perinatal lethality due to respiratory failure caused by a significant lung maturation delay. The few animals that reach adulthood have a normal lifespan, but present areas of atelectasis mixed with patches of emphysema and normal tissue in the lung. METHODS Eight double knockout and eight control mice were analyzed using micro-X-ray computerized tomography and a Small Animal Physiological Monitoring system. Tissues and samples from these mice were analyzed using standard histological and Molecular Biology methods and the significance of the results analyzed using a Student´s T-test. RESULTS The very few double knockout mice surviving up to adulthood display clear craniofacial abnormalities reminiscent of those seen in RASopathy mouse models, as well as thrombocytopenia, bleeding anomalies, and reduced platelet activation induced by thrombin. These surviving mice also present heart and spleen hyperplasia, and elevated numbers of myeloid-derived suppressor cells in the spleen. Mechanistically, we observed that these phenotypic alterations are accompanied by increased KRAS-GTP levels in heart, platelets and primary mouse embryonic fibroblasts from these animals. CONCLUSIONS Our data uncovers a new, previously unidentified mechanism capable of triggering a RASopathy phenotype in mice as a result of the combined removal of HRAS and NRAS.
Collapse
Affiliation(s)
- Rocío Fuentes-Mateos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
- Present address: Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, Netherlands
| | - Rósula García-Navas
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
| | - Cristina Fernández-Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Luis Hernández-Cano
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Present address: Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, Netherlands
| | - Nuria Calzada-Nieto
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
| | - Andrea Olarte-San Juan
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Eugenio Santos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain.
| | - Alberto Fernández-Medarde
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
9
|
Fasano G, Petrini S, Bonavolontà V, Paradisi G, Pedalino C, Tartaglia M, Lauri A. Assessment of the FRET-based Teen sensor to monitor ERK activation changes preceding morphological defects in a RASopathy zebrafish model and phenotypic rescue by MEK inhibitor. Mol Med 2024; 30:47. [PMID: 38594640 PMCID: PMC11005195 DOI: 10.1186/s10020-024-00807-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 03/12/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND RASopathies are genetic syndromes affecting development and having variable cancer predisposition. These disorders are clinically related and are caused by germline mutations affecting key players and regulators of the RAS-MAPK signaling pathway generally leading to an upregulated ERK activity. Gain-of-function (GOF) mutations in PTPN11, encoding SHP2, a cytosolic protein tyrosine phosphatase positively controlling RAS function, underlie approximately 50% of Noonan syndromes (NS), the most common RASopathy. A different class of these activating mutations occurs as somatic events in childhood leukemias. METHOD Here, we evaluated the application of a FRET-based zebrafish ERK reporter, Teen, and used quantitative FRET protocols to monitor non-physiological RASopathy-associated changes in ERK activation. In a multi-level experimental workflow, we tested the suitability of the Teen reporter to detect pan-embryo ERK activity correlates of morphometric alterations driven by the NS-causing Shp2D61G allele. RESULTS Spectral unmixing- and acceptor photobleaching (AB)-FRET analyses captured pathological ERK activity preceding the manifestation of quantifiable body axes defects, a morphological pillar used to test the strength of SHP2 GoF mutations. Last, the work shows that by multi-modal FRET analysis, we can quantitatively trace back the modulation of ERK phosphorylation obtained by low-dose MEK inhibitor treatment to early development, before the onset of morphological defects. CONCLUSION This work proves the usefulness of FRET imaging protocols on both live and fixed Teen ERK reporter fish to readily monitor and quantify pharmacologically- and genetically-induced ERK activity modulations in early embryos, representing a useful tool in pre-clinical applications targeting RAS-MAPK signaling.
Collapse
Affiliation(s)
- Giulia Fasano
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Stefania Petrini
- Microscopy facility, Research laboratories, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Valeria Bonavolontà
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Graziamaria Paradisi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
- Department for Innovation in Biological Agro-food and Forest systems (DIBAF), University of Tuscia, Viterbo, 01100, Italy
| | - Catia Pedalino
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy.
| | - Antonella Lauri
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, 00146, Italy.
| |
Collapse
|
10
|
Shiozawa Y, Fujita S, Nannya Y, Ogawa S, Nomura N, Kiguchi T, Sezaki N, Kudo H, Toyama T. First report of familial mixed phenotype acute leukemia: shared clinical characteristics, Philadelphia translocation, and germline variants. Int J Hematol 2024; 119:465-471. [PMID: 38424413 DOI: 10.1007/s12185-024-03724-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 03/02/2024]
Abstract
While our understanding of the molecular basis of mixed phenotype acute leukemia (MPAL) has progressed over the decades, our knowledge is limited and the prognosis remains poor. Investigating cases of familial leukemia can provide insights into the role of genetic and environmental factors in leukemogenesis. Although familial cases and associated mutations have been identified in some leukemias, familial occurrence of MPAL has never been reported. Here, we report the first cases of MPAL in a family. A 68-year-old woman was diagnosed with MPAL and received haploidentical stem cell transplantation from her 44-year-old son. In four years, the son himself developed MPAL. Both cases exhibited similar characteristics such as biphenotypic leukemia with B/myeloid cell antigens, Philadelphia translocation (BCR-ABL1 mutation), and response to acute lymphoblastic leukemia-type chemotherapy. These similarities suggest the presence of hereditary factors contributing to the development of MPAL. Targeted sequencing identified shared germline variants in these cases; however, in silico analyses did not strongly support their pathogenicity. Intriguingly, when the son developed MPAL, the mother did not develop donor-derived leukemia and remained in remission. Our cases provide valuable insights to guide future research on familial MPAL.
Collapse
Affiliation(s)
- Yuka Shiozawa
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan
| | - Shinya Fujita
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan.
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Division of Hematopoietic Disease Control, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Naho Nomura
- Department of Hematology, Chugoku Central Hospital of Japan Mutual Aid Association of Public School Teachers, Hiroshima, Japan
| | - Toru Kiguchi
- Saitama Medical Center, Department of Diabetes, Endocrinology and Hematology, Dokkyo Medical University, Saitama, Japan
| | - Nobuo Sezaki
- Department of Hematology, Chugoku Central Hospital of Japan Mutual Aid Association of Public School Teachers, Hiroshima, Japan
| | - Himari Kudo
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan
| | - Takaaki Toyama
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan
| |
Collapse
|
11
|
Cobaleda C, Godley LA, Nichols KE, Wlodarski MW, Sanchez-Garcia I. Insights into the Molecular Mechanisms of Genetic Predisposition to Hematopoietic Malignancies: The Importance of Gene-Environment Interactions. Cancer Discov 2024; 14:396-405. [PMID: 38426560 PMCID: PMC10913756 DOI: 10.1158/2159-8290.cd-23-1091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/20/2023] [Accepted: 12/01/2023] [Indexed: 03/02/2024]
Abstract
SUMMARY The recognition of host genetic factors underlying susceptibility to hematopoietic malignancies has increased greatly over the last decade. Historically, germline predisposition was thought to primarily affect the young. However, emerging data indicate that hematopoietic malignancies that develop in people of all ages across the human lifespan can derive from germline predisposing conditions and are not exclusively observed in younger individuals. The age at which hematopoietic malignancies manifest appears to correlate with distinct underlying biological pathways. Progression from having a deleterious germline variant to being diagnosed with overt malignancy involves complex, multistep gene-environment interactions with key external triggers, such as infection and inflammatory stimuli, driving clonal progression. Understanding the mechanisms by which predisposed clones transform under specific pressures may reveal strategies to better treat and even prevent hematopoietic malignancies from occurring.Recent unbiased genome-wide sequencing studies of children and adults with hematopoietic malignancies have revealed novel genes in which disease-causing variants are of germline origin. This paradigm shift is spearheaded by findings in myelodysplastic syndrome/acute myeloid leukemia (MDS/AML) as well as acute lymphoblastic leukemia, but it also encompasses other cancer types. Although not without challenges, the field of genetic cancer predisposition is advancing quickly, and a better understanding of the genetic basis of hematopoietic malignancies risk affects therapeutic decisions as well as genetic counseling and testing of at-risk family members.
Collapse
Affiliation(s)
- Cesar Cobaleda
- Immune System Development and Function Unit, Centro de Biología Molecular Severo Ochoa (CBM, CSIC-UAM), Madrid, Spain
| | - Lucy A. Godley
- Division of Hematology/Oncology, Department of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - Kim E. Nichols
- Division of Cancer Predisposition, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Marcin W. Wlodarski
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
12
|
Wachter F, Pikman Y. Pathophysiology of Acute Myeloid Leukemia. Acta Haematol 2024; 147:229-246. [PMID: 38228114 DOI: 10.1159/000536152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a biologically heterogenous disease arising in clonally proliferating hematopoietic stem cells. Sequential acquisition of mutations leads to expanded proliferation of clonal myeloid progenitors and failure of differentiation, leading to fulminant AML. SUMMARY Here, we review the pathophysiology of AML with a focus on factors predisposing to AML development, including prior chemo- and radiation therapy, environmental factors, and germline predisposition. KEY MESSAGE Increasing genomic characterization of AML and insight into mechanisms of its development will be critical to improvement in AML prognostication and therapy.
Collapse
Affiliation(s)
- Franziska Wachter
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Yana Pikman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Sakashita K, Yoshida N, Muramatsu H, Ohtsuka Y, Watanabe K, Yabe M, Kakuda H, Honda Y, Watanabe T, Haba M, Ohmori S, Matsuda K, Yuza Y, Saito A, Horibe K, Adachi S, Manabe A. Allogeneic Hematopoietic Cell Transplantation for Juvenile Myelomonocytic Leukemia with a Busulfan, Fludarabine, and Melphalan Regimen: JPLSG JMML-11. Transplant Cell Ther 2024; 30:105.e1-105.e10. [PMID: 37806448 DOI: 10.1016/j.jtct.2023.10.002] [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: 06/20/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML), which is classified as a myelodysplastic/myeloproliferative neoplasm, is a rare hematologic malignancy of childhood. Most patients with JMML require allogeneic hematopoietic cell transplantation (HCT) as a curative therapy. A Japanese retrospective analysis demonstrated favorable outcomes for a busulfan (BU) + fludarabine (FLU) + melphalan (MEL) regimen, with an overall survival (OS) of 72% and an event-free survival (EFS) of 53%. To further validate the efficacy and safety of this regimen, the Japan Pediatric Leukemia/Lymphoma Study Group (JPLSG) conducted a nationwide prospective study, JMML-11. Between July 2011 and June 2017, 28 patients with newly diagnosed JMML were enrolled in JMML11. Low-dose chemotherapy for tumor control before HCT was recommended, and patients treated with AML-type chemotherapy and azacitidine were excluded. The conditioning regimen comprised i.v. BU, 16 doses administered every 6 h, with dose adjustment based on pharmacokinetic (PK) studies on days -11 to -8; FLU, 30 mg/m2/day or 1 mg/kg/day for patients <10 kg or age <1 year on days -7 to -4; and MEL, 90 mg/m2/day or 3 mg/kg/day for patients <10 kg or <1 year on days -3 to -2. The donor was selected by the physician in charge. A family donor was available for 7 patients (3 HLA-matched siblings, 3 HLA-1-antigen mismatched parents, and 1 haploidentical father). Overall, 21 patients received grafts from unrelated donors, including 8 HLA-matched donors and 13 HLA-mismatched donors. The graft source was related bone marrow (BM) for 7 patients, unrelated BM for 14 patients, and unrelated cord blood for 7 patients. Neutrophil engraftment was achieved in 21 of 28 patients (75%), with a median of 20.5 days (range, 11 to 39 days) after transplantation. The 3-year OS, 3-year EFS, 3-year relapse rate, and 3-year transplantation-related mortality were 63% (95% confidence interval [CI], 42% to 78%), 52% (95% CI, 32% to 69%), 18% (95% CI, 6% to 34%), and 21% (95% CI, 9% to 38%), respectively. WBC count before the conditioning regimen (≥7.0 × 109/L) was significantly associated with inferior EFS and OS. Body surface area ≥.5 m2, spleen size <4 cm before conditioning, and HLA-matched unrelated BM donors were significantly associated with better OS. Adverse effects related to the conditioning regimen included febrile neutropenia (86%), diarrhea (39%), hypoxemia (21%), and mucositis (18%). BU-associated toxicity, including sinusoidal obstruction syndrome (SOS) and thrombotic microangiopathy (TMA), occurred in 7 patients (25%; SOS, n = 6; TMA, n = 2). Retrospective analysis of PK data after the first BU dose in 23 patients, including 6 with SOS and 17 without SOS, did not show significant differences between groups. The JMML-11 study confirms the positive results of previous retrospective analyses. BU+FLU+MEL might become a standard conditioning regimen for patients with JMML.
Collapse
Affiliation(s)
- Kazuo Sakashita
- Department of Hematology and Oncology, Nagano Children's Hospital, Azumino, Japan.
| | - Nao Yoshida
- Department of Hematology and Oncology, Children's Medical Center, Japanese Red Cross Aichi Medical Center Nagoya First Hospital, Nagoya, Japan
| | - Hideki Muramatsu
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Sizuoka, Japan
| | - Miharu Yabe
- Department of Innovative Medical Science, Tokai University School of Medicine, Isehara, Japan
| | - Harumi Kakuda
- Department of Hematology and Oncology, Chiba Children's Hospital, Chiba, Japan
| | - Yuko Honda
- Department of Pediatrics, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Tomoyuki Watanabe
- Department of Health and Nutritional Sciences, Faculty of Health Sciences, Aichi Gakuin University, Nisshin, Japan
| | - Masami Haba
- Department of Biopharmaceutics, Faculty of Pharmacy, Chiba Institute of Science, Choshi, Japan
| | - Shigeru Ohmori
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan
| | - Kazuyuki Matsuda
- Department of Clinical Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| | - Yuki Yuza
- Department of Pediatric Hematology Oncology, Tokyo Metropolitan Children's Medical Center, Fuchu, Japan
| | - Akiko Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Keizo Horibe
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Souichi Adachi
- Human Health Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
14
|
Pillay-Smiley N, Fletcher JS, de Blank P, Ratner N. Shedding New Light: Novel Therapies for Common Disorders in Children with Neurofibromatosis Type I. Pediatr Clin North Am 2023; 70:937-950. [PMID: 37704352 DOI: 10.1016/j.pcl.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Neurofibromatosis type I (NF1) is a common dominantly inherited disorder, and one of the most common of the RASopathies. Most individuals with NF1 develop plexiform neurofibromas and cutaneous neurofibromas, nerve tumors caused by NF1 loss of function in Schwann cells. Cell culture models and mouse models of NF1 are being used to test drug efficacy in preclinical trials, which led to Food and Drug Administration approval for use of MEK inhibitors to shrink most inoperable plexiform neurofibromas. This article details methods used for testing in preclinical models, and outlines newer models that may identify additional, curative, strategies.
Collapse
Affiliation(s)
- Natasha Pillay-Smiley
- University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jonathan S Fletcher
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Current Address: Division of Hematology-Oncology, University of Texas Southwestern, Dallas, TX, USA
| | - Peter de Blank
- University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-0731, USA; Cancer and Blood Diseases Institute, The Cure Starts Now Foundation Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
15
|
Reinig EF, Rubinstein JD, Patil AT, Schussman AL, Horner VL, Kanagal-Shamanna R, Churpek JE, Matson DR. Needle in a haystack or elephant in the room? Identifying germline predisposition syndromes in the setting of a new myeloid malignancy diagnosis. Leukemia 2023; 37:1589-1599. [PMID: 37393344 PMCID: PMC10529926 DOI: 10.1038/s41375-023-01955-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 06/03/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Myeloid malignancies associated with germline predisposition syndromes account for up to 10% of myeloid neoplasms. They are classified into three categories by the proposed 5th Edition of the World Health Organization Classification of Hematolymphoid Tumors: (1) neoplasms with germline predisposition without a pre-existing platelet disorder or organ dysfunction, (2) neoplasms with germline predisposition and pre-existing platelet disorder, or (3) neoplasms with germline predisposition and potential organ dysfunction. Recognizing these entities is critical because patients and affected family members benefit from interfacing with hematologists who specialize in these disorders and can facilitate tailored treatment strategies. However, identification of these syndromes in routine pathology practice is often challenging, as characteristic findings associated with these diagnoses at baseline are frequently absent, nonspecific, or impossible to evaluate in the setting of a myeloid malignancy. Here we review the formally classified germline predisposition syndromes associated with myeloid malignancies and summarize practical recommendations for pathologists evaluating a new myeloid malignancy diagnosis. Our intent is to empower clinicians to better screen for germline disorders in this common clinical setting. Recognizing when to suspect a germline predisposition syndrome, pursue additional ancillary testing, and ultimately recommend referral to a cancer predisposition clinic or hematology specialist, will ensure optimal patient care and expedite research to improve outcomes for these individuals.
Collapse
Affiliation(s)
- Erica F Reinig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Apoorva T Patil
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Amanda L Schussman
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Vanessa L Horner
- Wisconsin State Laboratory of Hygiene, University of Wisconsin-Madison, Madison, WI, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology and Molecular Diagnostics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jane E Churpek
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Blood Cancer Research Institute, Madison, WI, USA
| | - Daniel R Matson
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Wisconsin Blood Cancer Research Institute, Madison, WI, USA.
| |
Collapse
|
16
|
Patti G, Scaglione M, Maiorano NG, Rosti G, Divizia MT, Camia T, De Rose EL, Zucconi A, Casalini E, Napoli F, Di Iorgi N, Maghnie M. Abnormalities of pubertal development and gonadal function in Noonan syndrome. Front Endocrinol (Lausanne) 2023; 14:1213098. [PMID: 37576960 PMCID: PMC10422880 DOI: 10.3389/fendo.2023.1213098] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
Background Noonan syndrome (NS) is a genetic multisystem disorder characterised by variable clinical manifestations including dysmorphic facial features, short stature, congenital heart disease, renal anomalies, lymphatic malformations, chest deformities, cryptorchidism in males. Methods In this narrative review, we summarized the available data on puberty and gonadal function in NS subjects and the role of the RAS/mitogen-activated protein kinase (MAPK) signalling pathway in fertility. In addition, we have reported our personal experience on pubertal development and vertical transmission in NS. Conclusions According to the literature and to our experience, NS patients seem to have a delay in puberty onset compared to the physiological timing reported in healthy children. Males with NS seem to be at risk of gonadal dysfunction secondary not only to cryptorchidism but also to other underlying developmental factors including the MAP/MAPK pathway and genetics. Long-term data on a large cohort of males and females with NS are needed to better understand the impact of delayed puberty on adult height, metabolic profile and well-being. The role of genetic counselling and fertility related-issues is crucial.
Collapse
Affiliation(s)
- Giuseppa Patti
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Marco Scaglione
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Nadia Gabriella Maiorano
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Giulia Rosti
- Department of Clinical Genetics and Genomics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Maria Teresa Divizia
- Department of Clinical Genetics and Genomics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Tiziana Camia
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Elena Lucia De Rose
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Alice Zucconi
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Emilio Casalini
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Flavia Napoli
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Natascia Di Iorgi
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Mohamad Maghnie
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| |
Collapse
|
17
|
Sequeira V, Waddingham MT, Tsuchimochi H, Maack C, Pearson JT. Mechano-energetic uncoupling in hypertrophic cardiomyopathy: Pathophysiological mechanisms and therapeutic opportunities. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100036. [PMID: 39801694 PMCID: PMC11708264 DOI: 10.1016/j.jmccpl.2023.100036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 01/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a frequent inherited form of heart failure. The underlying cause of HCM is generally attributed to mutations in genes that encode for sarcomeric proteins, but the pathogenesis of the disease is also influenced by non-genetic factors, which can contribute to diastolic dysfunction and hypertrophic remodeling. Central to the pathogenesis of HCM is hypercontractility, a state that is an antecedent to several key derangements, including increased mitochondrial workload and oxidative stress. As a result, energy depletion and mechano-energetic uncoupling drive cardiac growth through signaling pathways such as ERK and/or potentially AMPK downregulation. Metabolic remodeling also occurs in HCM, characterized by decreased fatty acid oxidation and increased glucose uptake. In some instances, ketones may also feed the heart with energy and act as signaling molecules to reduce oxidative stress and hypertrophic signaling. In addition, arrhythmias are frequently triggered in HCM, resulting from the high Ca2+-buffering of the myofilaments and changes in the ATP/ADP ratio. Understanding the mechanisms driving the progression of HCM is critical to the development of effective therapeutic strategies. This paper presents evidence from both experimental and clinical studies that support the role of hypercontractility and cellular energy alterations in the progression of HCM towards heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Vasco Sequeira
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - Mark T. Waddingham
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
| | - Christoph Maack
- DZHI, Department of Translational Science Universitätsklinikum, Würzburg, Germany
| | - James T. Pearson
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita-shi, Osaka, Japan
- Department of Physiology and Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
18
|
Aukema SM, Glaser S, van den Hout MFCM, Dahlum S, Blok MJ, Hillmer M, Kolarova J, Sciot R, Schott DA, Siebert R, Stumpel CTRM. Molecular characterization of an embryonal rhabdomyosarcoma occurring in a patient with Kabuki syndrome: report and literature review in the light of tumor predisposition syndromes. Fam Cancer 2023; 22:103-118. [PMID: 35856126 PMCID: PMC9829644 DOI: 10.1007/s10689-022-00306-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/05/2022] [Indexed: 01/13/2023]
Abstract
Kabuki syndrome is a well-recognized syndrome characterized by facial dysmorphism and developmental delay/intellectual disability and in the majority of patients a germline variant in KMT2D is found. As somatic KMT2D variants can be found in 5-10% of tumors a tumor predisposition in Kabuki syndrome is discussed. So far less than 20 patients with Kabuki syndrome and a concomitant malignancy have been published. Here we report on a female patient with Kabuki syndrome and a c.2558_2559delCT germline variant in KMT2D who developed an embryonal rhabdomyosarcoma (ERMS) at 10 years. On tumor tissue we performed DNA-methylation profiling and exome sequencing (ES). Copy number analyses revealed aneuploidies typical for ERMS including (partial) gains of chromosomes 2, 3, 7, 8, 12, 15, and 20 and 3 focal deletions of chromosome 11p. DNA methylation profiling mapped the case to ERMS by a DNA methylation-based sarcoma classifier. Sequencing suggested gain of the wild-type KMT2D allele in the trisomy 12. Including our patient literature review identified 18 patients with Kabuki syndrome and a malignancy. Overall, the landscape of malignancies in patients with Kabuki syndrome was reminiscent of that of the pediatric population in general. Histopathological and molecular data were only infrequently reported and no report included next generation sequencing and/or DNA-methylation profiling. Although we found no strong arguments pointing towards KS as a tumor predisposition syndrome, based on the small numbers any relation cannot be fully excluded. Further planned studies including profiling of additional tumors and long term follow-up of KS-patients into adulthood could provide further insights.
Collapse
Affiliation(s)
- Sietse M Aukema
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - Selina Glaser
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Mari F C M van den Hout
- Department of Pathology, Research Institute GROW, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sonja Dahlum
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Marinus J Blok
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - Morten Hillmer
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Julia Kolarova
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Raf Sciot
- Department of Pathology, University Hospital, University of Leuven, 3000, Louvain, Belgium
| | - Dina A Schott
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands
- Department of Pediatrics, Zuyderland Medical Center, Heerlen, The Netherlands
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, Ulm, Germany
| | - Constance T R M Stumpel
- Department of Clinical Genetics, Maastricht University Medical Centre (MUMC+), PO Box 5800, 6202 AZ, Maastricht, The Netherlands.
- Department of Clinical Genetics and GROW-School for Oncology & Developmental Biology, Maastricht University Medical Center+, Maastricht, The Netherlands.
| |
Collapse
|
19
|
Unuma K, Tomomasa D, Noma K, Yamamoto K, Matsuyama TA, Makino Y, Hijikata A, Wen S, Ogata T, Okamoto N, Okada S, Ohashi K, Uemura K, Kanegane H. Case Report: Molecular autopsy underlie COVID-19-associated sudden, unexplained child mortality. Front Immunol 2023; 14:1121059. [PMID: 37143668 PMCID: PMC10151512 DOI: 10.3389/fimmu.2023.1121059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Herein, we report a child with COVID-19 and seemingly no underlying disease, who died suddenly. The autopsy revealed severe anemia and thrombocytopenia, splenomegaly, hypercytokinemia, and a rare ectopic congenital coronary origin. Immunohistochemical analysis demonstrated that the patient had acute lymphoblastic leukemia of the B-cell precursor phenotype (BCP-ALL). The complex cardiac and hematological abnormalities suggested the presence of an underlying disease; therefore, we performed whole-exome sequencing (WES). WES revealed a leucine-zipper-like transcription regulator 1 (LZTR1) variant, indicating Noonan syndrome (NS). Therefore, we concluded that the patient had underlying NS along with coronary artery malformation and that COVID-19 infection may have triggered the sudden cardiac death due to increased cardiac load caused by high fever and dehydration. In addition, multiple organ failure due to hypercytokinemia probably contributed to the patient's death. This case would be of interest to pathologists and pediatricians because of the limited number of NS patients with LZTR1 variants; the complex combination of an LZTR1 variant, BCP-ALL, and COVID-19; and a rare pattern of the anomalous origin of the coronary artery. Thus, we highlight the significance of molecular autopsy and the application of WES with conventional diagnostic methods.
Collapse
Affiliation(s)
- Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- *Correspondence: Kana Unuma,
| | - Dan Tomomasa
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kosuke Noma
- Department of Pediatrics, Hiroshima University, Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Taka-aki Matsuyama
- Department of Legal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Yohsuke Makino
- Department of Forensic Medicine, The University of Tokyo, Tokyo, Japan
| | - Atsushi Hijikata
- Department of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsutomu Ogata
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women’s and Children’s Hospital, Izumi, Osaka, Japan
| | - Satoshi Okada
- Department of Pediatrics, Hiroshima University, Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kenichi Ohashi
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
20
|
Burek M, Kaupp V, Blecharz-Lang K, Dilling C, Meybohm P. Protocadherin gamma C3: a new player in regulating vascular barrier function. Neural Regen Res 2023. [PMID: 35799511 PMCID: PMC9241426 DOI: 10.4103/1673-5374.343896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Defects in the endothelial cell barrier accompany diverse malfunctions of the central nervous system such as neurodegenerative diseases, stroke, traumatic brain injury, and systemic diseases such as sepsis, viral and bacterial infections, and cancer. Compromised endothelial sealing leads to leaking blood vessels, followed by vasogenic edema. Brain edema as the most common complication caused by stroke and traumatic brain injury is the leading cause of death. Brain microvascular endothelial cells, together with astrocytes, pericytes, microglia, and neurons form a selective barrier, the so-called blood-brain barrier, which regulates the movement of molecules inside and outside of the brain. Mechanisms that regulate blood-brain barrier permeability in health and disease are complex and not fully understood. Several newly discovered molecules that are involved in the regulation of cellular processes in brain microvascular endothelial cells have been described in the literature in recent years. One of these molecules that are highly expressed in brain microvascular endothelial cells is protocadherin gamma C3. In this review, we discuss recent evidence that protocadherin gamma C3 is a newly identified key player involved in the regulation of vascular barrier function.
Collapse
|
21
|
Zenker M, Edouard T, Blair JC, Cappa M. Noonan syndrome: improving recognition and diagnosis. Arch Dis Child 2022; 107:1073-1078. [PMID: 35246453 PMCID: PMC9685729 DOI: 10.1136/archdischild-2021-322858] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
Abstract
Noonan syndrome (NS) is a mostly dominantly inherited disorder affecting 1:1000 to 1:2500 live births. The phenotype varies in severity and can involve multiple organ systems over a patient's lifetime. Diagnosis is based on a combination of features, including typical facial features, short stature, skeletal abnormalities, presence of cardiac defects, mild developmental delay, cryptorchidism, lymphatic dysplasia and a family history of NS. The phenotype varies from oligosymptomatic adults without significant medical issues to severely affected neonates with life-threatening heart disease. Early, accurate diagnosis is important for individualised management and to optimise developmental and long-term outcomes, but mildly affected patients often go undiagnosed for both healthcare provider (HCP)-related and patient-related reasons. Lack of awareness of NS among HCPs means that some do not recognise the condition, particularly in mildly affected patients and families. Some families do not want to receive a diagnosis that medicalises a condition that may account for family traits (eg, distinctive facial features and short stature), particularly when a child's physical and cognitive development may be satisfactory. As for any condition with lifelong effects on multiple organ systems, a multidisciplinary approach provides the best care. It is proposed that increasing awareness of NS among non-specialist HCPs and other professionals could help direct a parent/carer to seek specialist advice and increase the number of NS diagnoses, with the potential to optimise lifelong patient outcomes. Non-specialists do not need to become experts in either diagnosis or treatment; however, early recognition of NS and referral to an appropriate specialist is important.
Collapse
Affiliation(s)
- Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Thomas Edouard
- Endocrine, Bone Diseases and Genetics Unit, Toulouse University Hospital, Toulouse, France
| | - Joanne C Blair
- Department of Paediatric Endocrinology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Marco Cappa
- Department of Endocrinology, Bambino Gesu Children's Hospital-Tor Vergata University, Rome, Italy
| |
Collapse
|
22
|
Ney G, Gross A, Livinski A, Kratz CP, Stewart DR. Cancer incidence and surveillance strategies in individuals with RASopathies. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:530-540. [PMID: 36533693 PMCID: PMC9825668 DOI: 10.1002/ajmg.c.32018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/15/2022] [Accepted: 11/20/2022] [Indexed: 12/24/2022]
Abstract
RASopathies are a set of clinical syndromes that have molecular and clinical overlap. Genetically, these syndromes are defined by germline pathogenic variants in RAS/MAPK pathway genes resulting in activation of this pathway. Clinically, their common molecular signature leads to comparable phenotypes, including cardiac anomalies, neurologic disorders and notably, elevated cancer risk. Cancer risk in individuals with RASopathies has been estimated from retrospective reviews and cohort studies. For example, in Costello syndrome, cancer incidence is significantly elevated over the general population, largely due to solid tumors. In some forms of Noonan syndrome, cancer risk is also elevated over the general population and is enriched for hematologic malignancies. Thus, cancer surveillance guidelines have been developed to monitor for the occurrence of such cancers in individuals with some RASopathies. These include abdominal ultrasound and urinalyses for individuals with Costello syndrome, while complete blood counts and splenic examination are recommended in Noonan syndrome. Improved cancer risk estimates and refinement of surveillance recommendations will improve the care of individuals with RASopathies.
Collapse
Affiliation(s)
- Gina Ney
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Maryland, USA
| | - Andrea Gross
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Alicia Livinski
- National Institutes of Health Library, National Institutes of Health, Bethesda, Maryland, USA
| | - Christian P Kratz
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, Maryland, USA
| |
Collapse
|
23
|
De Vos N, Hofmans M, Lammens T, De Wilde B, Van Roy N, De Moerloose B. Targeted therapy in juvenile myelomonocytic leukemia: Where are we now? Pediatr Blood Cancer 2022; 69:e29930. [PMID: 36094370 DOI: 10.1002/pbc.29930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/07/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare and aggressive clonal neoplasm of early childhood, classified as an overlap myeloproliferative/myelodysplastic neoplasm by the World Health Organization. In 90% of the patients with JMML, typical initiating mutations in the canonical Ras pathway genes NF1, PTPN11, NRAS, KRAS, and CBL can be identified. Hematopoietic stem cell transplantation (HSCT) currently is the established standard of care in most patients, although long-term survival is still only 50-60%. Given the limited therapeutic options and the important morbidity and mortality associated with HSCT, new therapeutic approaches are urgently needed. Hyperactivation of the Ras pathway as disease mechanism in JMML lends itself to the use of targeted therapy. Targeted therapy could play an important role in the future treatment of patients with JMML. This review presents a comprehensive overview of targeted therapies already developed and evaluated in vitro and in vivo in patients with JMML.
Collapse
Affiliation(s)
- Nele De Vos
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University, Ghent, Belgium
| | - Mattias Hofmans
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Tim Lammens
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bram De Wilde
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Nadine Van Roy
- Cancer Research Institute Ghent, Ghent, Belgium.,Center for Medical Genetics Ghent, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Abstract
OPINION STATEMENT Acute myeloid leukemia (AML) is the most common form of leukemia in adults, leading to the highest number of annual leukemia-associated deaths in the USA. Although most AML patients initially enter remission following induction therapy, most eventually relapse, underscoring the unmet need for more effective therapies. In recent years, novel high-throughput sequencing techniques, and mouse and human models of disease have increased our understanding of the molecular mechanisms that lead to AML. Leukemogenic mechanisms can be broadly classified into two types-cell-intrinsic and cell-extrinsic. Cell-intrinsic mechanisms include an array of genetic and epigenetic alterations that lead to dysregulated gene expression and function in hematopoietic stem/progenitor cells, leading to their increased fitness and ultimately, malignant transformation. Extrinsic mechanisms include both hematopoietic and non-hematopoietic stromal components of the leukemic microenvironment that interact with pre-leukemic and leukemic clones to promote their survival, self-renewal, and/or resistance to therapy. Through the individual and concerted action of these factors, pre-leukemic clones acquire the changes necessary for leukemic transformation. In addition, following therapy, specific leukemic clones are selected for that eventually re-initiate disease. Improving our understanding of these cell-intrinsic and cell-extrinsic mechanisms will provide novel opportunities to treat AML as well as prevent the development of disease.
Collapse
|
25
|
Saha A, Rai V, Kakoty S, Sawhney J, Kourav TPS. A case series of clinical & hematological profile of juvenile myelomonocytic leukemia(JMML). PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2022. [DOI: 10.1016/j.phoj.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
26
|
Stagi S, Ferrari V, Ferrari M, Priolo M, Tartaglia M. Inside the Noonan "universe": Literature review on growth, GH/IGF axis and rhGH treatment: Facts and concerns. Front Endocrinol (Lausanne) 2022; 13:951331. [PMID: 36060964 PMCID: PMC9434367 DOI: 10.3389/fendo.2022.951331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/18/2022] [Indexed: 12/21/2022] Open
Abstract
Noonan syndrome (NS) is a disorder characterized by a typical facial gestalt, congenital heart defects, variable cognitive deficits, skeletal defects, and short stature. NS is caused by germline pathogenic variants in genes coding proteins with a role in the RAS/mitogen-activated protein kinase signaling pathway, and it is typically associated with substantial genetic and clinical complexity and variability. Short stature is a cardinal feature in NS, with evidence indicating that growth hormone (GH) deficiency, partial GH insensitivity, and altered response to insulin-like growth factor I (IGF-1) are contributing events for growth failure in these patients. Decreased IGF-I, together with low/normal responses to GH pharmacological provocation tests, indicating a variable presence of GH deficiency/resistance, in particular in subjects with pathogenic PTPN11 variants, are frequently reported. Nonetheless, short- and long-term studies have demonstrated a consistent and significant increase in height velocity (HV) in NS children and adolescents treated with recombinant human GH (rhGH). While the overall experience with rhGH treatment in NS patients with short stature is reassuring, it is difficult to systematically compare published data due to heterogeneous protocols, potential enrolment bias, the small size of cohorts in many studies, different cohort selection criteria and varying durations of therapy. Furthermore, in most studies, the genetic information is lacking. NS is associated with a higher risk of benign and malignant proliferative disorders and hypertrophic cardiomyopathy, and rhGH treatment may further increase risk in these patients, especially as dosages vary widely. Herein we provide an updated review of aspects related to growth, altered function of the GH/IGF axis and cell response to GH/IGF stimulation, rhGH treatment and its possible adverse events. Given the clinical variability and genetic heterogeneity of NS, treatment with rhGH should be personalized and a conservative approach with judicious surveillance is recommended. Depending on the genotype, an individualized follow-up and close monitoring during rhGH treatments, also focusing on screening for neoplasms, should be considered.
Collapse
Affiliation(s)
- Stefano Stagi
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Vittorio Ferrari
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Marta Ferrari
- Department of Health Sciences, University of Florence, Anna Meyer Children’s University Hospital, Florence, Italy
| | - Manuela Priolo
- Medical Genetics Unit, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, Reggio Calabria, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
27
|
Symeonidis A, Chondropoulos S, Verigou E, Lazaris V, Kourakli A, Tsirigotis P. Allogeneic Hematopoietic Stem Cell Transplantation for Mixed or Overlap Myelodysplastic/Myeloproliferative Disorders. Front Oncol 2022; 12:884723. [PMID: 35992818 PMCID: PMC9389581 DOI: 10.3389/fonc.2022.884723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022] Open
Abstract
Chronic myelomonocytic leukemia (CMML) and the remaining, less frequent hybrid, mixed, or overlap myelodysplastic syndromes/myeloproliferative neoplasms (MDSs/MPNs) are difficult to treat neoplastic hematological disorders, exhibiting substantial clinical and prognostic heterogeneity, for which clear therapeutic guidelines or effective treatment options are still missing. CMML has an overall survival ranging from a few months to several years. Although patients with proliferative or dysplastic features may benefit from hydroxyurea and hypomethylating agent treatment, respectively, none of these treatments can establish long-term remission and prevent the inevitable transformation to acute leukemia. Novel targeted treatment approaches are emerging but are still under investigation. Therefore, currently, allogeneic stem cell transplantation (allo-SCT) remains the only treatment modality with a curative potential, but its widespread application is limited, due to significant morbidity and mortality associated with the procedure, especially in the elderly and in patients with comorbidities. Recognition of patient eligibility for allo-SCT is crucial, and the procedure should be addressed to patients with a good performance status without severe comorbidities and mainly to those in intermediate- to high-risk category, with a suitable stem cell donor available. The issues of best timing for performing transplantation, patient and donor eligibility, the type of conditioning regimen, and the outcomes after various allo-SCT procedures are the topics of this review.
Collapse
Affiliation(s)
- Argiris Symeonidis
- University of Patras Medical School, Hematology Division, Patras, Greece
- *Correspondence: Argiris Symeonidis, ; orcid.org/0000-0002-0543-046X
| | | | - Evgenia Verigou
- Hematology Division, General University Hospital of Patras, Rion of Patras, Greece
| | - Vasileios Lazaris
- Hematology Division, General University Hospital of Patras, Rion of Patras, Greece
| | - Alexandra Kourakli
- Hematology Division, General University Hospital of Patras, Rion of Patras, Greece
| | - Panagiotis Tsirigotis
- Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
28
|
Genomic and Epigenomic Landscape of Juvenile Myelomonocytic Leukemia. Cancers (Basel) 2022; 14:cancers14051335. [PMID: 35267643 PMCID: PMC8909150 DOI: 10.3390/cancers14051335] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myelodysplastic/myeloproliferative neoplasm characterized by the constitutive activation of the RAS pathway. In spite of the recent progresses in the molecular characterization of JMML, this disease is still a clinical challenge due to its heterogeneity, difficult diagnosis, poor prognosis, and the lack of curative treatment options other than hematopoietic stem cell transplantation (HSCT). In this review, we will provide a detailed overview of the genetic and epigenetic alterations occurring in JMML, and discuss their clinical relevance in terms of disease prognosis and risk of relapse after HSCT. We will also present the most recent advances on novel preclinical and clinical therapeutic approaches directed against JMML molecular targets. Finally, we will outline future research perspectives to further explore the oncogenic mechanism driving JMML leukemogenesis and progression, with special attention to the application of single-cell next-generation sequencing technologies. Abstract Juvenile myelomonocytic leukemia (JMML) is a rare myelodysplastic/myeloproliferative neoplasm of early childhood. Most of JMML patients experience an aggressive clinical course of the disease and require hematopoietic stem cell transplantation, which is currently the only curative treatment. JMML is characterized by RAS signaling hyperactivation, which is mainly driven by mutations in one of five genes of the RAS pathway, including PTPN11, KRAS, NRAS, NF1, and CBL. These driving mutations define different disease subtypes with specific clinico-biological features. Secondary mutations affecting other genes inside and outside the RAS pathway contribute to JMML pathogenesis and are associated with a poorer prognosis. In addition to these genetic alterations, JMML commonly presents aberrant epigenetic profiles that strongly correlate with the clinical outcome of the patients. This observation led to the recent publication of an international JMML stratification consensus, which defines three JMML clinical groups based on DNA methylation status. Although the characterization of the genomic and epigenomic landscapes in JMML has significantly contributed to better understand the molecular mechanisms driving the disease, our knowledge on JMML origin, cell identity, and intratumor and interpatient heterogeneity is still scarce. The application of new single-cell sequencing technologies will be critical to address these questions in the future.
Collapse
|
29
|
Role of CBL Mutations in Cancer and Non-Malignant Phenotype. Cancers (Basel) 2022; 14:cancers14030839. [PMID: 35159106 PMCID: PMC8833995 DOI: 10.3390/cancers14030839] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/30/2022] Open
Abstract
Simple Summary CBL mutations are progressively being described as involved in different clinical manifestations. Somatic CBL mutations can be found in different type of cancer. The clinical spectrum of germline mutations configures the so-called CBL syndrome, a cancer-predisposing condition that includes multisystemic involvement characterized by variable phenotypic expression and expressivity. In this review we provide an up-to-date review of the clinical manifestation of CBL mutations and of the molecular mechanisms in which CBL exerts its pathogenic role. Abstract CBL plays a key role in different cell pathways, mainly related to cancer onset and progression, hematopoietic development and T cell receptor regulation. Somatic CBL mutations have been reported in a variety of malignancies, ranging from acute myeloid leukemia to lung cancer. Growing evidence have defined the clinical spectrum of germline CBL mutations configuring the so-called CBL syndrome; a cancer-predisposing condition that also includes multisystemic involvement characterized by variable phenotypic expression and expressivity. This review provides a comprehensive overview of the molecular mechanisms in which CBL exerts its function and describes the clinical manifestation of CBL mutations in humans.
Collapse
|
30
|
Szczawińska-Popłonyk A, Popłonyk N, Niedziela M, Sowińska-Seidler A, Sztromwasser P, Jamsheer A, Obara-Moszyńska M. Case report: The cardio-facio-cutaneous syndrome due to a novel germline mutation in MAP2K1: A multifaceted disease with immunodeficiency and short stature. Front Pediatr 2022; 10:990111. [PMID: 36313893 PMCID: PMC9614356 DOI: 10.3389/fped.2022.990111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022] Open
Abstract
Cardio-facio-cutaneous syndrome (CFCS) belongs to the group of RASopathies, clinical disorders defined by disruptions in the RAS/MAPK signaling pathway. It is caused by heterozygous gain-of-function germline mutations in genes encoding protein kinases: BRAF, MAP2K1 (MEK1), MAP2K2 (MEK2), and in the GTPase-encoding gene KRAS. CFCS is characterized by craniofacial dysmorphic features, congenital heart defects, severe malnutrition, proportionate short stature, anomalies within the structure of skin and hair, and psychomotor disability. The pathophysiology of growth impairment is multifactorial with feeding difficulties, growth hormone deficiency, and insensitivity. Immunodeficiency has not been hitherto reported as an integral part of CFCS yet an increased activation of the RAS/MAPK signaling pathway may contribute to explaining the causal relationship between RASopathy and the dysfunctions within the B and T lymph cell compartments resulting in a deficiency in T cell costimulation and B cell maturation with impaired class switch recombination, somatic hypermutation, and high-affinity antibody production. We report on a boy born prematurely at 32 WGA, with the perinatal period complicated by pneumonia, respiratory distress syndrome, and valvular pulmonary stenosis. The boy suffered from recurrent pneumonia, obstructive bronchitis, sepsis, urinary tract infection, and recurrent fevers. He presented with severe hypotrophy, psychomotor disability, short stature, craniofacial dysmorphism, dental hypoplasia, sparse hair, and cryptorchidism. Whole genome sequencing showed a novel heterozygous pathogenic germline missense variant: c.364A > G; p.Asn122Asp in the MAP2K1 gene, supporting the diagnosis of CFCS. The immunological workup revealed hypogammaglobulinemia, IgG subclass, and specific antibody deficiency accompanied by decreased numbers of T helper cells and naive and memory B cells. Replacement immunoglobulin therapy with timely antibiotic prophylaxis were instituted. At the age of six years, growth hormone deficiency was diagnosed and the rGH therapy was started. The ever-increasing progress in genetic studies contributes to establishing the definitive CFCS diagnosis and sheds the light on the interrelated genotype-phenotype heterogeneity of RASopathies. Herein, we add new phenotypic features of predominating humoral immunodeficiency to the symptomatology of CFCS with a novel mutation in MAP2K1. While CFCS is a multifaceted disease, increased pediatricians' awareness is needed to prevent the delay in diagnostics and therapeutic interventions.
Collapse
Affiliation(s)
- Aleksandra Szczawińska-Popłonyk
- Department of Pediatric Pneumonology, Allergy and Clinical Immunology, Institute of Pediatrics, Poznań University of Medical Sciences, Poznań, Poland
| | - Natalia Popłonyk
- Student Scientific Society for Pediatric Endocrinology, Poznań University of Medical Sciences, Poznań, Poland
| | - Marek Niedziela
- Department of Pediatric Endocrinology and Rheumatology, Institute of Pediatrics, Poznań University of Medical Sciences, Poznań, Poland
| | - Anna Sowińska-Seidler
- Department of Medical Genetics, Poznań University of Medical Sciences, Poznań, Poland
| | - Paweł Sztromwasser
- Department of Biostatistics and Translational Medicine, Medical University of Łódź, Łódź, Poland.,MNM Diagnostics, Poznań, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznań University of Medical Sciences, Poznań, Poland
| | - Monika Obara-Moszyńska
- Department of Pediatric Endocrinology and Rheumatology, Institute of Pediatrics, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
31
|
Huang S, Zhao G, Wu J, Li K, Wang Q, Fu Y, Zhang H, Bi Q, Li X, Wang W, Guo C, Zhang D, Wu L, Li X, Xu H, Han M, Wang X, Lei C, Qiu X, Li Y, Li J, Dai P, Yuan Y. Gene4HL: An Integrated Genetic Database for Hearing Loss. Front Genet 2021; 12:773009. [PMID: 34733322 PMCID: PMC8558372 DOI: 10.3389/fgene.2021.773009] [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: 09/09/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
Hearing loss (HL) is one of the most common disabilities in the world. In industrialized countries, HL occurs in 1–2/1,000 newborns, and approximately 60% of HL is caused by genetic factors. Next generation sequencing (NGS) has been widely used to identify many candidate genes and variants in patients with HL, but the data are scattered in multitudinous studies. It is a challenge for scientists, clinicians, and biologists to easily obtain and analyze HL genes and variant data from these studies. Thus, we developed a one-stop database of HL-related genes and variants, Gene4HL (http://www.genemed.tech/gene4hl/), making it easy to catalog, search, browse and analyze the genetic data. Gene4HL integrates the detailed genetic and clinical data of 326 HL-related genes from 1,608 published studies, along with 62 popular genetic data sources to provide comprehensive knowledge of candidate genes and variants associated with HL. Additionally, Gene4HL supports the users to analyze their own genetic engineering network data, performs comprehensive annotation, and prioritizes candidate genes and variations using custom parameters. Thus, Gene4HL can help users explain the function of HL genes and the clinical significance of variants by correlating the genotypes and phenotypes in humans.
Collapse
Affiliation(s)
- Shasha Huang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Guihu Zhao
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, China
| | - Jie Wu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Kuokuo Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, China
| | - Qiuquan Wang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Ying Fu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Honglei Zhang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Qingling Bi
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Xiaohong Li
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Weiqian Wang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Chang Guo
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Dejun Zhang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Lihua Wu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Xiaoge Li
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Huiyan Xu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Mingyu Han
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Xin Wang
- Angen Gene Medicine Technology Co., Ltd., Beijing, China
| | - Chen Lei
- Angen Gene Medicine Technology Co., Ltd., Beijing, China
| | - Xiaofang Qiu
- Angen Gene Medicine Technology Co., Ltd., Beijing, China
| | - Yang Li
- Angen Gene Medicine Technology Co., Ltd., Beijing, China
| | - Jinchen Li
- National Clinical Research Center for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China.,Center for Medical Genetics & Hunan Key Laboratory, School of Life Sciences, Central South University, Changsha, China
| | - Pu Dai
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Yongyi Yuan
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China.,National Clinical Research Center for Otolaryngologic Diseases, State Key Lab of Hearing Science, Ministry of Education, Beijing, China.,Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| |
Collapse
|
32
|
St. Martin EC, Ferrer A, Wudhikarn K, Mangaonkar A, Hogan W, Tefferi A, Gangat N, Lasho T, Altman JK, Godley LA, Patnaik MM. Clinical features and survival outcomes in patients with chronic myelomonocytic leukemia arising in the context of germline predisposition syndromes. Am J Hematol 2021; 96:E327-E330. [PMID: 34028844 DOI: 10.1002/ajh.26250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022]
Affiliation(s)
| | - Alejandro Ferrer
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | - Kitsada Wudhikarn
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | - Abhishek Mangaonkar
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | - William Hogan
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | - Ayalew Tefferi
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | - Naseema Gangat
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | - Terra Lasho
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| | | | - Lucy A. Godley
- Section of Hematology‐Oncology, Departments of Medicine and Human Genetics The University of Chicago Chicago Illinois USA
| | - Mrinal M. Patnaik
- Division of Hematology, Department of Internal Medicine Mayo Clinic Rochester Minnesota USA
| |
Collapse
|
33
|
Nathany S, Chatterjee G, Ghai S, Moulik NR, Shetty D, Subramanian PG, Tembhare P, Gujral S, Dhamne C, Banavali S, Narula G, Patkar N. Mutational landscape of Juvenile Myelomonocytic Leukemia (JMML)-A real-world context. Int J Lab Hematol 2021; 43:1531-1538. [PMID: 34387930 DOI: 10.1111/ijlh.13680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/22/2021] [Accepted: 07/31/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Juvenile myelomonocytic leukemia (JMML) is a rare childhood neoplasm (<5% cases), which has been categorized under myelodysplastic/myeloproliferative neoplasms (MDS/MPN) in the recent classification by the World Health Organization. METHODS We developed a 51-gene (151.5kB) low-cost targeted myeloid panel based on single-molecule molecular inversion probes to comprehensively evaluate the genomic profile of Juvenile myelomonocytic leukemia (JMML). RESULTS A total of 50 children with clinical and pathological features of JMML were sequenced at high coverage. Among the 50 patients, 44(88%) harbored mutations in one of the RAS/MAPK-pathway genes, most frequently in NRAS (32%), followed by PTPN11 (28%) and NF1 (22%). One-fifth of children had more than one mutation, with 5 cases harboring two RAS pathway mutations. Monosomy 7 was detected in 32% (16) patients, and five of these did not harbor any RAS pathway mutations. Children with monosomy 7 showed shorter overall survival compared with their wild-type counterparts (P = .02). CONCLUSION Our study highlights that comprehensive genomic profiling identifies at least one mutation in almost 90% of JMML patients. Performing genomic analysis at baseline might help in triaging children with JMML for allogenic stem cell transplant in resource-constrained settings.
Collapse
Affiliation(s)
- Shrinidhi Nathany
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Gaurav Chatterjee
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Shruti Ghai
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India
| | - Nirmalya Roy Moulik
- Homi Bhabha National Institute (HBNI), Mumbai, India.,Pediatric Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Dhanalaxmi Shetty
- Homi Bhabha National Institute (HBNI), Mumbai, India.,Department of Cancer Cytogenetics, Advanced Centre for treatment and research in cancer, Tata Memorial Centre, Mumbai, India
| | - P G Subramanian
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Prashant Tembhare
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Sumeet Gujral
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Chetan Dhamne
- Homi Bhabha National Institute (HBNI), Mumbai, India.,Pediatric Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Sripad Banavali
- Homi Bhabha National Institute (HBNI), Mumbai, India.,Pediatric Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Gaurav Narula
- Homi Bhabha National Institute (HBNI), Mumbai, India.,Pediatric Haematolymphoid Disease Management Group, Tata Memorial Centre, Mumbai, India
| | - Nikhil Patkar
- Department of Hematopathology, Advanced Centre for Treatment and Research in Cancer, Tata Memorial Centre, Mumbai, India.,Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
34
|
Mariani RA, Jennings L, Zhang S, Bhat R, Gong S. Morphologic and Immunophenotypic Differences in Juvenile Myelomonocytic Leukemias With CBL and Other Canonical RAS-pathway Gene Mutations: A Single Institutional Experience. J Pediatr Hematol Oncol 2021; 43:e819-e825. [PMID: 33769390 DOI: 10.1097/mph.0000000000002149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/22/2021] [Indexed: 11/25/2022]
Abstract
The diagnostic criteria for juvenile myelomonocytic leukemia have recently been revised to include clinical findings and RAS-pathway gene mutations per the 2016 World Health Organization Classification of Tumors of Hematopoietic and Lymphoid Tissues. Differing clinical behaviors have been observed in cases with CBL versus other RAS-pathway gene (RAS-p) mutations, notably the patients with CBL mutations can be self-limiting with spontaneous resolution. Additional clinical characteristics and histopathologic findings between these subsets are less well-described. We performed a retrospective search and identified cases with either CBL or RAS-p mutations, as per targeted and/or massively parallel sequencing. Eight patients had sufficient material for review, including cytogenetic studies and peripheral blood, bone marrow aspirate, and/or biopsy with flow cytometry analyses. Three patients showed CBL mutations and lower percentages of hemoglobin F and peripheral blood absolute monocyte counts, lesser degrees of leukocytosis compared with the RAS-p cohort, and normal megakaryocyte morphology and myeloblast immunophenotypes. Two of these patients were managed with observation only and experienced resolution of their disease. The patients with RAS-p mutations had severe thrombocytopenia, moderate to severe anemia, and experienced variable clinical outcomes. Abnormal megakaryocyte morphology and decreased numbers of megakaryocytes were seen in cases with RAS-p mutations. In addition, 3 of 4 cases with flow cytometry data demonstrated aberrant CD7 expression in myeloblasts. Our study is the first to identify morphologic and immunophenotypic differences between juvenile myelomonocytic leukemia cases with CBL or RAS-p mutations, and further supports previous reports of significantly different clinical behaviors between these subsets of patients.
Collapse
Affiliation(s)
| | | | - Shanxiang Zhang
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN
| | - Rukhmi Bhat
- Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, Ann and Robert H. Lurie Children's Hospital, Chicago, IL
| | | |
Collapse
|
35
|
Mayerhofer C, Niemeyer CM, Flotho C. Current Treatment of Juvenile Myelomonocytic Leukemia. J Clin Med 2021; 10:3084. [PMID: 34300250 PMCID: PMC8305558 DOI: 10.3390/jcm10143084] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare pediatric leukemia characterized by mutations in five canonical RAS pathway genes. The diagnosis is made by typical clinical and hematological findings associated with a compatible mutation. Although this is sufficient for clinical decision-making in most JMML cases, more in-depth analysis can include DNA methylation class and panel sequencing analysis for secondary mutations. NRAS-initiated JMML is heterogeneous and adequate management ranges from watchful waiting to allogeneic hematopoietic stem cell transplantation (HSCT). Upfront azacitidine in KRAS patients can achieve long-term remissions without HSCT; if HSCT is required, a less toxic preparative regimen is recommended. Germline CBL patients often experience spontaneous resolution of the leukemia or exhibit stable mixed chimerism after HSCT. JMML driven by PTPN11 or NF1 is often rapidly progressive, requires swift HSCT and may benefit from pretransplant therapy with azacitidine. Because graft-versus-leukemia alloimmunity is central to cure high risk patients, the immunosuppressive regimen should be discontinued early after HSCT.
Collapse
Affiliation(s)
- Christina Mayerhofer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
| | - Charlotte M. Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (C.M.); (C.M.N.)
- German Cancer Consortium (DKTK), 79106 Freiburg, Germany
| |
Collapse
|
36
|
RASopathies: from germline mutations to somatic and multigenic diseases. Biomed J 2021; 44:422-432. [PMID: 34175492 PMCID: PMC8514848 DOI: 10.1016/j.bj.2021.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
The RAS-RAF-MEK-ERK signaling pathway is vital for different cellular mechanisms including cell proliferation, differentiation and apoptosis. This importance is highlighted by the high prevalence of mutations in RAS or related proteins of the pathway in cancers. More recently, development abnormalities have been linked to various germline mutations in this pathway and called RASopathies. Interestingly, rare disorders such as RAS-associated leukoproliferative diseases and histiocytosis have also been recently linked to multiple mutations in the same pathway, sometimes with the same mutation. This review will focus on germline RASopathies and rare somatic RASopathies and focus on how gain-of-function mutations in the same pathway can lead to various diseases.
Collapse
|
37
|
Implications of mosaicism in variant interpretation: A case of a de novo homozygous NF1 variant. Eur J Med Genet 2021; 64:104236. [PMID: 33965620 DOI: 10.1016/j.ejmg.2021.104236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/21/2021] [Accepted: 04/28/2021] [Indexed: 11/23/2022]
Abstract
Neurofibromatosis type 1 is a common multisystem autosomal dominant syndrome caused by pathogenic heterozygous variants in the neurofibromin gene (NF1). It is associated with a substantially increased cancer risk. Mosaicism for NF1 has been clinically well-established for "second hit" variants in skin lesions and tumor tissues. Here, we report on a 3-month-old boy with multiple café au lait macules (CAMs) and juvenile myelomonocytic leukemia (JMML) who was found to carry a previously established pathogenic NF1 variant (c.586+5G>A), as revealed by whole-exome sequencing. Surprisingly, however, this variant was detected in the homozygous state in the patient and was absent in the parents and siblings. Deep sequencing of this variant using blood, buccal swabs and skin samples was performed. As expected for an NF1 gene mutation promoting JMML, the variant was detected in 90.6% of the blood DNA reads, in sharp contrast to the mere 5% and 0.74% of reads in the saliva- and skin fibroblast-derived DNA, respectively. Our analysis, therefore, confirmed postzygotic origin of the variant followed by a mitotic event resulting in its homozygosity, although we could not differentiate between the possibilities of a gene conversion and mitotic crossover. Apparently de novo homozygous variants should trigger a careful investigation into mosaicism to achieve accurate interpretation.
Collapse
|
38
|
Hasserjian RP, Buckstein R, Patnaik MM. Navigating Myelodysplastic and Myelodysplastic/Myeloproliferative Overlap Syndromes. Am Soc Clin Oncol Educ Book 2021; 41:328-350. [PMID: 34010050 DOI: 10.1200/edbk_320113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Myelodysplastic syndromes (MDS) and MDS/myeloproliferative neoplasms (MPNs) are clonal diseases that differ in morphologic diagnostic criteria but share some common disease phenotypes that include cytopenias, propensity to acute myeloid leukemia evolution, and a substantially shortened patient survival. MDS/MPNs share many clinical and molecular features with MDS, including frequent mutations involving epigenetic modifier and/or spliceosome genes. Although the current 2016 World Health Organization classification incorporates some genetic features in its diagnostic criteria for MDS and MDS/MPNs, recent accumulation of data has underscored the importance of the mutation profiles on both disease classification and prognosis. Machine-learning algorithms have identified distinct molecular genetic signatures that help refine prognosis and notable associations of these genetic signatures with morphologic and clinical features. Combined geno-clinical models that incorporate mutation data seem to surpass the current prognostic schemes. Future MDS classification and prognostication schema will be based on the portfolio of genetic aberrations and traditional features, such as blast count and clinical factors. Arriving at these systems will require studies on large patient cohorts that incorporate advanced computational analysis. The current treatment algorithm in MDS is based on patient risk as derived from existing prognostic and disease classes. Luspatercept is newly approved for patients with MDS and ring sideroblasts who are transfusion dependent after erythropoietic-stimulating agent failure. Other agents that address red blood cell transfusion dependence in patients with lower-risk MDS and the failure of hypomethylating agents in higher-risk disease are in advanced testing. Finally, a plethora of novel targeted agents and immune checkpoint inhibitors are being evaluated in combination with a hypomethylating agent backbone to augment the depth and duration of response and, we hope, improve overall survival.
Collapse
Affiliation(s)
| | - Rena Buckstein
- Division of Hematology/Oncology, Sunnybrook Odette Cancer Center, Toronto, Ontario, Canada
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, MN
| |
Collapse
|
39
|
Mitchell SG, Pencheva B, Westfall E, Porter CC. Cancer Predisposition in Neonates and Infants: Recognition, Tumor Types, and Surveillance. Clin Perinatol 2021; 48:1-14. [PMID: 33583498 DOI: 10.1016/j.clp.2020.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Pediatric cancer is rare, and malignancy during the neonatal period even rarer. However, several malignancies can present in infancy, most commonly in the form of solid tumors. Specific cancer types, bilateral or multifocal disease, associated congenital malformations, and/or cancers in close relatives may herald a diagnosis of an underlying cancer predisposition syndrome. For many patients, surveillance protocols are recommended beginning at birth or during the course of maternal prenatal care. Advantages and disadvantages of genetic testing and surveillance should be discussed with families using a multidisciplinary approach, with input from a genetic counselor with expertise in pediatric cancer predisposition.
Collapse
Affiliation(s)
- Sarah G Mitchell
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Bojana Pencheva
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Ellie Westfall
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA
| | - Christopher C Porter
- Department of Pediatrics, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, 1760 Haygood Drive, Atlanta, GA 30322, USA.
| |
Collapse
|
40
|
Greenmyer JR, Kohorst M. Pediatric Neoplasms Presenting with Monocytosis. Curr Hematol Malig Rep 2021; 16:235-246. [PMID: 33630234 DOI: 10.1007/s11899-021-00611-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW Juvenile myelomonocytic leukemia (JMML) is a rare but severe pediatric neoplasm with hematopoietic stem cell transplant as its only established curative option. The development of targeted therapeutics for JMML is being guided by an understanding of the pathobiology of this condition. Here, we review JMML with an emphasis on genetics in order to (i) demonstrate the relationship between JMML genotype and clinical phenotype and (ii) explore potential genetic targets of novel JMML therapies. RECENT FINDINGS DNA hypermethylation studies have demonstrated consistently that methylation is related to disease severity. Increasing understanding of methylation in JMML may open the door to novel therapies, such as DNA methyltransferase inhibitors. The PI3K/AKT/MTOR, JAK/STAT, and RAF/MEK/ERK pathways are being investigated as therapeutic targets for JMML. Future therapy for JMML will be driven by an increased understanding of pathobiology. Targeted therapeutic approaches hold potential for improving outcomes in patients with JMML.
Collapse
Affiliation(s)
| | - Mira Kohorst
- Pediatric Hematology and Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
41
|
Gupta AK, Meena JP, Chopra A, Tanwar P, Seth R. Juvenile myelomonocytic leukemia-A comprehensive review and recent advances in management. AMERICAN JOURNAL OF BLOOD RESEARCH 2021; 11:1-21. [PMID: 33796386 PMCID: PMC8010610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/24/2021] [Indexed: 06/12/2023]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myelodysplastic/myeloproliferative neoplasm overlap disease. JMML is associated with mutations in the RAS pathway genes resulting in the myeloid progenitors being sensitive to granulocyte monocyte colony-stimulating factor (GM-CSF). Karyotype abnormalities and additional epigenetic alterations can also be found in JMML. Neurofibromatosis and Noonan's syndrome have a predisposition for JMML. In a few patients, the RAS genes (NRAS, KRAS, and PTPN11) are mutated at the germline and this usually results in a transient myeloproliferative disorder with a good prognosis. JMML with somatic RAS mutation behaves aggressively. JMML presents with cytopenias and leukemic infiltration into organs. The laboratory findings include hyperleukocytosis, monocytosis, increased hemoglobin-F levels, and circulating myeloid precursors. The blast cells in the peripheral blood/bone-marrow aspirate are less than 20% and the absence of the BCR-ABL translocation helps to differentiate from chronic myeloid leukemia. JMML should be differentiated from immunodeficiencies, viral infections, intrauterine infections, hemophagolymphohistiocytosis, other myeloproliferative disorders, and leukemias. Chemotherapy is employed as a bridge to HSCT, except in few with less aggressive disease, in which chemotherapy alone can result in long term remission. Azacitidine has shown promise as a single agent to stabilize the disease. The prognosis of JMML is poor with about 50% of patients surviving after an allogeneic hematopoietic stem cell transplant (HSCT). Allogeneic HSCT is the only known cure for JMML to date. Myeloablative conditioning is most commonly used with graft versus host disease (GVHD) prophylaxis tailored to the aggressiveness of the disease. Relapses are common even after HSCT and a second HSCT can salvage a third of these patients. Novel options in the treatment of JMML e.g., hypomethylating agents, MEK inhibitors, JAK inhibitors, tyrosine kinase inhibitors, etc. are being explored.
Collapse
Affiliation(s)
- Aditya Kumar Gupta
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Jagdish Prasad Meena
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Anita Chopra
- Laboratory Oncology Unit, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Pranay Tanwar
- Laboratory Oncology Unit, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical SciencesNew Delhi 110029, India
| | - Rachna Seth
- Division of Pediatric Oncology, Department of Pediatrics, All India Institute of Medical SciencesNew Delhi 110029, India
| |
Collapse
|
42
|
Molecular Targeted Therapy in Myelodysplastic Syndromes: New Options for Tailored Treatments. Cancers (Basel) 2021; 13:cancers13040784. [PMID: 33668555 PMCID: PMC7917605 DOI: 10.3390/cancers13040784] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Myelodysplastic syndromes (MDS) are a group of diseases in which bone marrow stem cells acquire genetic alterations and can initiate leukemia, blocking the production of mature blood cells. It is of crucial importance to identify those genetic abnormalities because some of them can be the targeted. To date only very few drugs are approved for patients manifesting this group of disorders and there is an urgent need to develop new effective therapies. This review gives an overview of the genetic of MDS and the therapeutic options available and in clinical experimentation. Abstract Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoietic disorders characterized by ineffective hematopoiesis, progressive cytopenias and increased risk of transformation to acute myeloid leukemia. The improved understanding of the underlying biology and genetics of MDS has led to better disease and risk classification, paving the way for novel therapeutic opportunities. Indeed, we now have a vast pipeline of targeted agents under pre-clinical and clinical development, potentially able to modify the natural history of the diverse disease spectrum of MDS. Here, we review the latest therapeutic approaches (investigational and approved agents) for MDS treatment. A deep insight will be given to molecularly targeted therapies by reviewing new agents for individualized precision medicine.
Collapse
|
43
|
Hofmans M, Lammens T, Depreter B, Wu Y, Erlacher M, Caye A, Cavé H, Flotho C, de Haas V, Niemeyer CM, Stary J, Van Nieuwerburgh F, Deforce D, Van Loocke W, Van Vlierberghe P, Philippé J, De Moerloose B. Long non-coding RNAs as novel therapeutic targets in juvenile myelomonocytic leukemia. Sci Rep 2021; 11:2801. [PMID: 33531590 PMCID: PMC7854679 DOI: 10.1038/s41598-021-82509-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) treatment primarily relies on hematopoietic stem cell transplantation and results in long-term overall survival of 50-60%, demonstrating a need to develop novel treatments. Dysregulation of the non-coding RNA transcriptome has been demonstrated before in this rare and unique disorder of early childhood. In this study, we investigated the therapeutic potential of targeting overexpressed long non-coding RNAs (lncRNAs) in JMML. Total RNA sequencing of bone marrow and peripheral blood mononuclear cell preparations from 19 untreated JMML patients and three healthy children revealed 185 differentially expressed lncRNA genes (131 up- and 54 downregulated). LNA GapmeRs were designed for 10 overexpressed and validated lncRNAs. Molecular knockdown (≥ 70% compared to mock control) after 24 h of incubation was observed with two or more independent GapmeRs in 6 of them. For three lncRNAs (lnc-THADA-4, lnc-ACOT9-1 and NRIR) knockdown resulted in a significant decrease of cell viability after 72 h of incubation in primary cultures of JMML mononuclear cells, respectively. Importantly, the extent of cellular damage correlated with the expression level of the lncRNA of interest. In conclusion, we demonstrated in primary JMML cell cultures that knockdown of overexpressed lncRNAs such as lnc-THADA-4, lnc-ACOT9-1 and NRIR may be a feasible therapeutic strategy.
Collapse
Affiliation(s)
- Mattias Hofmans
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium. .,Department of Diagnostic Sciences, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium.
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Barbara Depreter
- Department of Laboratory Medicine Hematology, University Hospital Brussels, Brussels, Belgium
| | - Ying Wu
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Miriam Erlacher
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg, German Cancer Research Center, Heidelberg, Germany
| | - Aurélie Caye
- Department of Genetics, University Hospital of Robert Debré (APHP) and INSERM U1131, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Hélène Cavé
- Department of Genetics, University Hospital of Robert Debré (APHP) and INSERM U1131, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg, German Cancer Research Center, Heidelberg, Germany
| | - Valerie de Haas
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Dutch Childhood Oncology Group, The Hague, The Netherlands
| | - Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg, German Cancer Research Center, Heidelberg, Germany
| | - Jan Stary
- Department of Pediatric Hematology/Oncology, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Filip Van Nieuwerburgh
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Dieter Deforce
- Laboratory for Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Wouter Van Loocke
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Pieter Van Vlierberghe
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| |
Collapse
|
44
|
Dal Molin A, Hofmans M, Gaffo E, Buratin A, Cavé H, Flotho C, de Haas V, Niemeyer CM, Stary J, Van Vlierberghe P, Philippé J, De Moerloose B, Te Kronnie G, Bresolin S, Lammens T, Bortoluzzi S. CircRNAs Dysregulated in Juvenile Myelomonocytic Leukemia: CircMCTP1 Stands Out. Front Cell Dev Biol 2021; 8:613540. [PMID: 33490078 PMCID: PMC7815690 DOI: 10.3389/fcell.2020.613540] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML), a rare myelodysplastic/myeloproliferative neoplasm of early childhood, is characterized by clonal growth of RAS signaling addicted stem cells. JMML subtypes are defined by specific RAS pathway mutations and display distinct gene, microRNA (miRNA) and long non-coding RNA expression profiles. Here we zoom in on circular RNAs (circRNAs), molecules that, when abnormally expressed, may participate in malignant deviation of cellular processes. CirComPara software was used to annotate and quantify circRNAs in RNA-seq data of a “discovery cohort” comprising 19 JMML patients and 3 healthy donors (HD). In an independent set of 12 JMML patients and 6 HD, expression of 27 circRNAs was analyzed by qRT-PCR. CircRNA-miRNA-gene networks were reconstructed using circRNA function prediction and gene expression data. We identified 119 circRNAs dysregulated in JMML and 59 genes showing an imbalance of the circular and linear products. Our data indicated also circRNA expression differences among molecular subgroups of JMML. Validation of a set of deregulated circRNAs in an independent cohort of JMML patients confirmed the down-regulation of circOXNAD1 and circATM, and a marked up-regulation of circLYN, circAFF2, and circMCTP1. A new finding in JMML links up-regulated circMCTP1 with known tumor suppressor miRNAs. This and other predicted interactions with miRNAs connect dysregulated circRNAs to regulatory networks. In conclusion, this study provides insight into the circRNAome of JMML and paves the path to elucidate new molecular disease mechanisms putting forward circMCTP1 up-regulation as a robust example.
Collapse
Affiliation(s)
- Anna Dal Molin
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Mattias Hofmans
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| | - Enrico Gaffo
- Department of Molecular Medicine, University of Padova, Padua, Italy
| | - Alessia Buratin
- Department of Molecular Medicine, University of Padova, Padua, Italy.,Department of Biology, University of Padova, Padua, Italy
| | - Hélène Cavé
- Department of Genetics, University Hospital of Robert Debré, Paris, France.,INSERM U1131, Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Valerie de Haas
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Dutch Childhood Oncology Group, The Hague, Netherlands
| | - Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Stary
- Department of Pediatric Hematology/Oncology, Charles University and University Hospital Motol, Prague, Czechia
| | - Pieter Van Vlierberghe
- Cancer Research Institute Ghent, Ghent, Belgium.,Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | | | - Silvia Bresolin
- Onco-Hematology, Stem Cell Transplant and Gene Therapy Laboratory, IRP-Istituto di Ricerca Pediatrica, Padua, Italy.,Department of Maternal and Child Health, Padua University, Padua, Italy
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Stefania Bortoluzzi
- Department of Molecular Medicine, University of Padova, Padua, Italy.,Interdepartmental Research Center for Innovative Biotechnologies, University of Padova, Padua, Italy
| |
Collapse
|
45
|
Patnaik MM, Lasho TL. Genomics of myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:450-459. [PMID: 33275756 PMCID: PMC7727543 DOI: 10.1182/hematology.2020000130] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN) overlap syndromes are uniquely classified neoplasms occurring in both children and adults. This category consists of 5 neoplastic subtypes: chronic myelomonocytic leukemia (CMML), juvenile myelomonocytic leukemia (JMML), BCR-ABL1-negative atypical chronic myeloid leukemia (aCML), MDS/MPN-ring sideroblasts and thrombocytosis (MDS/MPN-RS-T), and MDS/MPN-unclassifiable (U). Cytogenetic abnormalities and somatic copy number variations are uncommon; however, >90% patients harbor gene mutations. Although no single gene mutation is specific to a disease subtype, certain mutational signatures in the context of appropriate clinical and morphological features can be used to establish a diagnosis. In CMML, mutated coexpression of TET2 and SRSF2 results in clonal hematopoiesis skewed toward monocytosis, and the ensuing acquisition of driver mutations including ASXL1, NRAS, and CBL results in overt disease. MDS/MPN-RS-T demonstrates features of SF3B1-mutant MDS with ring sideroblasts (MDS-RS), with the development of thrombocytosis secondary to the acquisition of signaling mutations, most commonly JAK2V617F. JMML, the only pediatric entity, is a bona fide RASopathy, with germline and somatic mutations occurring in the oncogenic RAS pathway giving rise to disease. BCR-ABL1-negative aCML is characterized by dysplastic neutrophilia and is enriched in SETBP1 and ETNK1 mutations, whereas MDS/MPN-U is the least defined and lacks a characteristic mutational signature. Molecular profiling also provides prognostic information, with truncating ASXL1 mutations being universally detrimental and germline CBL mutations in JMML showing spontaneous regression. Sequencing information in certain cases can help identify potential targeted therapies (IDH1, IDH2, and splicing mutations) and should be a mainstay in the diagnosis and management of these neoplasms.
Collapse
Affiliation(s)
- Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Terra L Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
46
|
Wong JC, Perez-Mancera PA, Huang TQ, Kim J, Grego-Bessa J, Del Pilar Alzamora M, Kogan SC, Sharir A, Keefe SH, Morales CE, Schanze D, Castel P, Hirose K, Huang GN, Zenker M, Sheppard D, Klein OD, Tuveson DA, Braun BS, Shannon K. KrasP34R and KrasT58I mutations induce distinct RASopathy phenotypes in mice. JCI Insight 2020; 5:140495. [PMID: 32990679 PMCID: PMC7710308 DOI: 10.1172/jci.insight.140495] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/24/2020] [Indexed: 01/16/2023] Open
Abstract
Somatic KRAS mutations are highly prevalent in many cancers. In addition, a distinct spectrum of germline KRAS mutations causes developmental disorders called RASopathies. The mutant proteins encoded by these germline KRAS mutations are less biochemically and functionally activated than those in cancer. We generated mice harboring conditional KrasLSL-P34Rand KrasLSL-T58I knock-in alleles and characterized the consequences of each mutation in vivo. Embryonic expression of KrasT58I resulted in craniofacial abnormalities reminiscent of those seen in RASopathy disorders, and these mice exhibited hyperplastic growth of multiple organs, modest alterations in cardiac valvulogenesis, myocardial hypertrophy, and myeloproliferation. By contrast, embryonic KrasP34R expression resulted in early perinatal lethality from respiratory failure due to defective lung sacculation, which was associated with aberrant ERK activity in lung epithelial cells. Somatic Mx1-Cre–mediated activation in the hematopoietic compartment showed that KrasP34R and KrasT58I expression had distinct signaling effects, despite causing a similar spectrum of hematologic diseases. These potentially novel strains are robust models for investigating the consequences of expressing endogenous levels of hyperactive K-Ras in different developing and adult tissues, for comparing how oncogenic and germline K-Ras proteins perturb signaling networks and cell fate decisions, and for performing preclinical therapeutic trials. Mouse models are developed to accurately recapitulate multiple features of RASopathy disorders caused by germline KRASP34R and KRAST581 mutations.
Collapse
Affiliation(s)
- Jasmine C Wong
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Pedro A Perez-Mancera
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Tannie Q Huang
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Jangkyung Kim
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Joaquim Grego-Bessa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Maria Del Pilar Alzamora
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | | | - Amnon Sharir
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California, USA
| | - Susan H Keefe
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California, USA
| | - Carolina E Morales
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Denny Schanze
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Pau Castel
- Helen Diller Family Comprehensive Cancer Center
| | - Kentaro Hirose
- Cardiovascular Research Institute.,Department of Physiology, and
| | - Guo N Huang
- Cardiovascular Research Institute.,Department of Physiology, and
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Magdeburg, Germany
| | - Dean Sheppard
- Cardiovascular Research Institute.,Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Ophir D Klein
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA.,Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, California, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA.,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, New York, USA
| | - Benjamin S Braun
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
47
|
Neven Q, Boulanger C, Bruwier A, de Ville de Goyet M, Meyts I, Moens L, Van Damme A, Brichard B. Clinical Spectrum of Ras-Associated Autoimmune Leukoproliferative Disorder (RALD). J Clin Immunol 2020; 41:51-58. [PMID: 33011939 DOI: 10.1007/s10875-020-00883-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/26/2020] [Indexed: 12/30/2022]
Abstract
Ras-associated autoimmune leukoproliferative disorder (RALD) is a clinical entity initially identified in patients evaluated for an autoimmune lymphoproliferative syndrome (ALPS)-like phenotype. It remains a matter of debate whether RALD is a chronic and benign lymphoproliferative disorder or a pre-malignant condition. We report the case of a 7-year-old girl diagnosed with RALD due to somatic KRAS mutation who progressed to a juvenile myelomonocytic leukemia phenotype and finally evolved into acute myeloid leukemia. The case report prompted a literature review by a search for all RALD cases published in PubMed and Embase. We identified 27 patients with RALD. The male-to-female ratio was 1:1 and median age at disease onset was 2 years (range 3 months-36 years). Sixteen patients (59%) harbored somatic mutations in KRAS and 11 patients (41%) somatic mutations in NRAS. The most common features were splenomegaly (26/27 patients), autoimmune cytopenia (15/16 patients), monocytosis (18/24 patients), pericarditis (6 patients), and skin involvement (4 patients). Two patients went on to develop a hematopoietic malignancy. In summary, the current case documents an additional warning about the long-term risk of malignancy in RALD.
Collapse
Affiliation(s)
- Quentin Neven
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| | - Cécile Boulanger
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Annelyse Bruwier
- Department of Pediatrics, Grand Hôpital de Charleroi, Charleroi, Belgium
| | - Maëlle de Ville de Goyet
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Isabelle Meyts
- Laboratory for Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, University Hospitals Leuven, Leuven, Belgium
- Department of Pediatrics, ERN-RITA Core Center, University Hospitals Leuven, Leuven, Belgium
| | - Leen Moens
- Laboratory for Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - An Van Damme
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Bénédicte Brichard
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| |
Collapse
|
48
|
Tseu B, Siow W, Pushkaran B, Cheesebrough B, Bain BJ. Beta thalassemia major and Noonan syndrome - Two genetic disorders manifest in the blood film. Am J Hematol 2020; 95:1113-1114. [PMID: 32112432 DOI: 10.1002/ajh.25767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 02/27/2020] [Indexed: 11/07/2022]
Affiliation(s)
- Bing Tseu
- Department of Haematology, Buckinghamshire Healthcare NHS Trust, Amersham, UK
| | - Wenchee Siow
- Department of Haematology, Buckinghamshire Healthcare NHS Trust, Amersham, UK
| | - Beena Pushkaran
- Department of Haematology, Buckinghamshire Healthcare NHS Trust, Amersham, UK
| | - Beth Cheesebrough
- Department of Paediatrics, Buckinghamshire Healthcare NHS Trust, Amersham, UK
| | - Barbara J Bain
- Department of Haematology, St Mary's Hospital Campus of Imperial College Faculty of Medicine, St Mary's Hospital, London, UK
| |
Collapse
|
49
|
Germline predisposition in myeloid neoplasms: Unique genetic and clinical features of GATA2 deficiency and SAMD9/SAMD9L syndromes. Best Pract Res Clin Haematol 2020; 33:101197. [PMID: 33038986 PMCID: PMC7388796 DOI: 10.1016/j.beha.2020.101197] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Increasing awareness about germline predisposition and the widespread application of unbiased whole exome sequencing contributed to the discovery of new clinical entities with high risk for the development of haematopoietic malignancies. The revised 2016 WHO classification introduced a novel category of "myeloid neoplasms with germline predisposition" with GATA2, CEBPA, DDX41, RUNX1, ANKRD26 and ETV6 genes expanding the spectrum of hereditary myeloid neoplasms (MN). Since then, more germline causes of MN were identified, including SAMD9, SAMD9L, and ERCC6L2. This review describes the genetic and clinical spectrum of predisposition to MN. The main focus lies in delineation of phenotypes, genetics and management of GATA2 deficiency and the novel SAMD9/SAMD9L-related disorders. Combined, GATA2 and SAMD9/SAMD9L (SAMD9/9L) syndromes are recognized as most frequent causes of primary paediatric myelodysplastic syndromes, particularly in setting of monosomy 7. To date, ~550 cases with germline GATA2 mutations, and ~130 patients with SAMD9/9L mutations had been reported in literature. GATA2 deficiency is a highly penetrant disorder with a progressive course that often rapidly necessitates bone marrow transplantation. In contrast, SAMD9/9L disorders show incomplete penetrance with various clinical outcomes ranging from spontaneous haematological remission observed in young children to malignant progression.
Collapse
|
50
|
Gao X, Huang SS, Qiu SW, Su Y, Wang WQ, Xu HY, Xu JC, Kang DY, Dai P, Yuan YY. Congenital sensorineural hearing loss as the initial presentation of PTPN11-associated Noonan syndrome with multiple lentigines or Noonan syndrome: clinical features and underlying mechanisms. J Med Genet 2020; 58:465-474. [PMID: 32737134 DOI: 10.1136/jmedgenet-2020-106892] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/22/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Germline variants in PTPN11 are the primary cause of Noonan syndrome with multiple lentigines (NSML) and Noonan syndrome (NS), which share common skin and facial symptoms, cardiac anomalies and retardation of growth. Hearing loss is considered an infrequent feature in patients with NSML/NS. However, in our cohort, we identified a group of patients with PTPN11 pathogenic variants that were primarily manifested in congenital sensorineural hearing loss (SNHL). This study evaluated the incidence of PTPN11-related NSML or NS in patients with congenital SNHL and explored the expression of PTPN11 and the underlying mechanisms in the auditory system. METHODS A total of 1502 patients with congenital SNHL were enrolled. Detailed phenotype-genotype correlations were analysed in patients with PTPN11 variants. Immunolabelling of Ptpn11 was performed in P35 mice. Zebrafish with Ptpn11 knockdown/mutant overexpression were constructed to further explore mechanism underlying the phenotypes. RESULTS Ten NSML/NS probands were diagnosed via the identification of pathogenic variants of PTPN11, which accounted for ~0.67% of the congenital SNHL cases. In mice cochlea, Shp2, which is encoded by Ptpn11, is distributed in the spiral ganglion neurons, hair cells and supporting cells of the inner ear. In zebrafish, knockdown of ptpn11a and overexpression of mutant PTPN11 were associated with a significant decrease in hair cells and supporting cells. We concluded that congenital SNHL could be a major symptom in PTPN11-associated NSML or NS. Other features may be mild, especially in children. CONCLUSION Screening for PTPN11 in patients with congenital hearing loss and variant-based diagnoses are recommended.
Collapse
Affiliation(s)
- Xue Gao
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
- Department of Otolaryngology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Sha-Sha Huang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Shi-Wei Qiu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Yu Su
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Wei-Qian Wang
- Department of Otolaryngology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Hui-Yan Xu
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Jin-Cao Xu
- Department of Otolaryngology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Dong-Yang Kang
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Pu Dai
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| | - Yong-Yi Yuan
- College of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital; National Clinical Research Center for Otolaryngologic Diseases; State Key Lab of Hearing Science, Ministry of Education; Beijing Key Lab of Hearing Impairment Prevention and Treatment, Beijing, China
| |
Collapse
|