51
|
Ziogas IA, Kakos CD, Wu WK, Montenovo MI, Matsuoka LK, Zarnegar-Lumley S, Alexopoulos SP. Liver Transplantation for Langerhans Cell Histiocytosis: A US Population-Based Analysis and Systematic Review of the Literature. Liver Transpl 2021; 27:1181-1190. [PMID: 33484600 DOI: 10.1002/lt.25995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/22/2020] [Accepted: 01/07/2021] [Indexed: 12/12/2022]
Abstract
Langerhans cell histiocytosis (LCH) is the most common histiocytic disorder. Liver involvement is seen in 10.1% to 19.8% of patients with LCH and can lead to secondary sclerosing cholangitis requiring liver transplantation (LT). We describe the characteristics and outcomes of patients undergoing LT for LCH. All patients undergoing a first LT for LCH in the United States were identified in the Scientific Registry of Transplant Recipients (SRTR) database (1987-2018). The Kaplan-Meier curve method and log-rank tests evaluated post-LT survival. A systematic literature review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) statement. A total of 60 LCH LT recipients were identified in the SRTR, and 55 patients (91.7%) were children with median total bilirubin levels at LT of 5.8 mg/dL (interquartile range [IQR], 2.7-12.9). A total of 49 patients (81.7%) underwent deceased donor LT (DDLT). The 1-year, 3-year, and 5-year patient survival rates were 86.6%, 82.4%, and 82.4%, respectively. The systematic review yielded 26 articles reporting on 50 patients. Of the patients, 41 were children (82.0%), 90.0% had multisystem LCH, and most patients underwent DDLT (91.9%; n = 34/37). Pre-LT chemotherapy was administered in 74.0% and steroids in 71.7% (n = 33/46) of the patients, and a recurrence of LCH to the liver was reported in 8.0% of the patients. Of the 50 patients, 11 (22.0%) died during a median follow-up of 25.2 months (IQR, 9.0-51.6), and the 1-year patient survival rate was 79.4%. LT can be considered as a feasible life-saving option for the management of liver failure secondary to LCH in well-selected patients.
Collapse
Affiliation(s)
- Ioannis A Ziogas
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN.,Surgery Working Group, Society of Junior Doctors, Athens, Greece
| | - Christos D Kakos
- Surgery Working Group, Society of Junior Doctors, Athens, Greece
| | - W Kelly Wu
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN
| | - Martin I Montenovo
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN
| | - Lea K Matsuoka
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN
| | - Sara Zarnegar-Lumley
- Department of Pediatrics, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN
| | - Sophoclis P Alexopoulos
- Department of Surgery, Division of Hepatobiliary Surgery and Liver Transplantation, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
52
|
Emile JF, Cohen-Aubart F, Collin M, Fraitag S, Idbaih A, Abdel-Wahab O, Rollins BJ, Donadieu J, Haroche J. Histiocytosis. Lancet 2021; 398:157-170. [PMID: 33901419 PMCID: PMC9364113 DOI: 10.1016/s0140-6736(21)00311-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Histiocytoses constitute a heterogeneous group of rare disorders, characterised by infiltration of almost any organ by myeloid cells with diverse macrophage or dendritic cell phenotypes. Histiocytoses can start at any age. Diagnosis is based on histology in combination with appropriate clinical and radiological findings. The low incidence and broad spectrum of clinical manifestations often leads to diagnostic delay, especially for adults. In most cases, biopsy specimens infiltrated by histiocytes have somatic mutations in genes activating the MAP kinase cell-signalling pathway. These mutations might also be present in blood cells and haematopoietic progenitors of patients with multisystem disease. A comprehensive range of investigations and molecular typing are essential to accurately predict prognosis, which can vary from spontaneous resolution to life-threatening disseminated disease. Targeted therapies with BRAF or MEK inhibitors have revolutionised salvage treatment. However, the type and duration of treatment are still debated, and the prevention of neurological sequelae remains a crucial issue.
Collapse
Affiliation(s)
- Jean-François Emile
- EA4340 BECCOH, Université de Versailles SQY, Service de Pathologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France.
| | - Fleur Cohen-Aubart
- Internal Medicine Department 2, French National Referral Center for Rare Systemic Diseases and Histiocytoses, Pitié-Salpêtrière Hospital, AP-HP and Sorbonne Université, Paris, France
| | - Matthew Collin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sylvie Fraitag
- Pathology Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Ahmed Idbaih
- UMR S 1127, CNRS/Inserm, Institut du Cerveau et de la Moelle Épinière, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, AP-HP and Sorbonne Université, Paris, France
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Barrett J Rollins
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean Donadieu
- EA4340 BECCOH, Université de Versailles SQY, Service de Pathologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France; Service d'Hématologie Oncologie Pédiatrique, Centre de Référence des Histiocytoses, Hôpital Armand-Trousseau, AP-HP, Paris, France
| | - Julien Haroche
- Internal Medicine Department 2, French National Referral Center for Rare Systemic Diseases and Histiocytoses, Pitié-Salpêtrière Hospital, AP-HP and Sorbonne Université, Paris, France
| |
Collapse
|
53
|
Jain P, Surrey LF, Straka J, Russo P, Womer R, Li MM, Storm PB, Waanders AJ, Hogarty MD, Resnick AC, Picarsic J. BRAF fusions in pediatric histiocytic neoplasms define distinct therapeutic responsiveness to RAF paradox breakers. Pediatr Blood Cancer 2021; 68:e28933. [PMID: 33565241 DOI: 10.1002/pbc.28933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/12/2021] [Indexed: 01/26/2023]
Abstract
Pediatric histiocytic neoplasms are hematopoietic disorders frequently driven by the BRAF-V600E mutation. Here, we identified two BRAF gene fusions (novel MTAP-BRAF and MS4A6A-BRAF) in two aggressive histiocytic neoplasms. In contrast to previously described BRAF fusions, MTAP-BRAF and MS4A6A-BRAF do not respond to the paradox breaker RAF inhibitor (RAFi) PLX8394 due to stable fusion dimerization mediated by the N-terminal fusion partners. This highlights a significant and clinically relevant shift from the current dogma that BRAF-fusions respond similarly to BRAF-inhibitors. As an alternative, we show suppression of fusion-driven oncogenic growth with the pan-RAFi LY3009120 and MEK inhibition.
Collapse
Affiliation(s)
- Payal Jain
- Center for Data Driven Discovery in Biomedicine (D3B), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lea F Surrey
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joshua Straka
- Center for Data Driven Discovery in Biomedicine (D3B), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Pierre Russo
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Richard Womer
- Department of Pediatrics, Division of Hematology and Oncology, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Marilyn M Li
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Phillip B Storm
- Center for Data Driven Discovery in Biomedicine (D3B), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurosurgery, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Angela J Waanders
- Department of Pediatrics, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA
| | - Michael D Hogarty
- Department of Pediatrics, Division of Hematology and Oncology, Children's Hospital of Philadelphia (CHOP), Philadelphia, Pennsylvania, USA
| | - Adam C Resnick
- Center for Data Driven Discovery in Biomedicine (D3B), Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jennifer Picarsic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
54
|
Tzotzola V, Petrikkos L, Papadakis V, Mitropoulou G, Kelaidi C, Dimitriadis E, Polychronopoulou S. Long-term outcome, clinical course and treatment approaches of paediatric langerhans cell histiocytosis: A greek reference centre report. Acta Paediatr 2021; 110:1944-1951. [PMID: 33382132 DOI: 10.1111/apa.15743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
AIM Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia with diverse clinical behaviour. In this article, we studied the clinical course, management and long-term outcomes of a paediatric cohort treated by our reference centre. METHODS We retrospectively studied 66 children with LCH, consecutively diagnosed by a Greek reference centre from 1974 to 2020. RESULTS The patients had a median age of 3.9 (range 0.0-15.9) years, 39 and 6 patients were diagnosed with unifocal or multifocal single system disease and 14 and 7 had multisystem disease with or without risk organ involvement. No late occurrence of clinical neurodegenerative disease or diabetes insipidus were observed at a median follow-up period of 4.1 (range 0.5-27.7) years. The 10-year event-free survival and overall survival were 65.0% and 90.3% and improved significantly over a 45-year period. Survival was superior in single system than multisystem cases. BRAF V600E mutation was found in 8/14 tested patients. Reactivation occurred in 12/66 patients (18.2%); 11 achieved remission and one patient died after a second relapse. CONCLUSION LCH survival rates significantly increased in our cohort over time. Reactivation occurred in 18.2% patients, but no late neurodegeneration was found. The prognostic value of single system disease status vs. multisystem LCH was confirmed.
Collapse
Affiliation(s)
- Vasiliki Tzotzola
- Department of Pediatric Hematology‐Oncology “Aghia Sophia” Children’s Hospital Athens Greece
| | - Loizos Petrikkos
- Department of Pediatric Hematology‐Oncology “Aghia Sophia” Children’s Hospital Athens Greece
| | - Vassilios Papadakis
- Department of Pediatric Hematology‐Oncology “Aghia Sophia” Children’s Hospital Athens Greece
| | | | - Charikleia Kelaidi
- Department of Pediatric Hematology‐Oncology “Aghia Sophia” Children’s Hospital Athens Greece
| | | | - Sophia Polychronopoulou
- Department of Pediatric Hematology‐Oncology “Aghia Sophia” Children’s Hospital Athens Greece
| |
Collapse
|
55
|
Fu Z, Li H, Arslan ME, Ells PF, Lee H. Hepatic Langerhans cell histiocytosis: A review. World J Clin Oncol 2021; 12:335-341. [PMID: 34131565 PMCID: PMC8173326 DOI: 10.5306/wjco.v12.i5.335] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/19/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatic Langerhans cell histiocytosis (LCH) is characterized by proliferation and accumulation of Langerhans cells in the liver, causing liver dysfunction or forming a mass lesion. The liver can be involved in isolation, or be affected along with other organs. A common clinical hepatic presentation is cholestasis with pruritis, fatigue and direct hyperbilirubinemia. In late stages, there may be hypoalbuminemia. Liver biopsy may be required for the diagnosis of hepatic LCH. Histologic finding may be diverse, including lobular Langerhans cell infiltrate with mixed inflammatory background, primary biliary cholangitis-like pattern, sclerosing cholangitis-like pattern, and even cirrhosis at later stages. Because of its non-specific injury patterns with broad differential diagnosis, establishing a diagnosis of hepatic LCH can be challenging. Hepatic LCH can easily be missed unless this diagnosis is considered at the time of biopsy interpretation. A definitive diagnosis relies on positive staining with CD1a and S100 antigen. Liver involvement is a high risk feature in LCH. The overall prognosis of hepatic LCH is poor. Treating at an early stage may improve the outcome. Systemic chemotherapy is the mainstay of treatment and liver transplantation may be offered. New molecular markers involved in pathogenesis of LCH are being explored with a potential for targeted therapy. However, further studies are needed to improve outcome.
Collapse
Affiliation(s)
- Zhiyan Fu
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY 12208, United States
| | - Hua Li
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY 12208, United States
| | - Mustafa Erdem Arslan
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY 12208, United States
| | - Peter F Ells
- Department of Gastroenterology, Albany Medical Center, Albany, NY 12208, United States
| | - Hwajeong Lee
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY 12208, United States
| |
Collapse
|
56
|
BRAFV 600E or mutant MAP2K1 human CD34+ cells establish Langerhans cell-like histiocytosis in immune-deficient mice. Blood Adv 2021; 4:4912-4917. [PMID: 33035332 DOI: 10.1182/bloodadvances.2020001926] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/30/2020] [Indexed: 12/14/2022] Open
Abstract
Key Points
BRAFV600E or mutant MAP2K1 expression in human CB CD34+ HSPCs lead to Langerhans cell–like histiocytosis in immune-deficient mice. BRAFV600E-expressing human CB CD34+ HSPCs did not generate hairy cell leukemia in xenograft mouse models.
Collapse
|
57
|
Massoth LR, Hung YP, Ferry JA, Hasserjian RP, Nardi V, Nielsen GP, Sadigh S, Venkataraman V, Selig M, Friedmann AM, Samore W, Killian JK, Milante R, Giessinger J, Foley-Peres K, Marcus C, Severson E, Duncan D, Sivakumar S, Ross JS, Desphande V, Ramkissoon SH, Vergilio JA, Louissaint A, Zukerberg LR, Williams EA. Histiocytic and Dendritic Cell Sarcomas of Hematopoietic Origin Share Targetable Genomic Alterations Distinct from Follicular Dendritic Cell Sarcoma. Oncologist 2021; 26:e1263-e1272. [PMID: 33904632 PMCID: PMC8265357 DOI: 10.1002/onco.13801] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Background Histiocytic and dendritic cell neoplasms are a diverse group of tumors arising from monocytic or dendritic cell lineage. Whereas the genomic features for Langerhans cell histiocytosis and Erdheim‐Chester disease have been well described, other less common and often aggressive tumors in this broad category remain poorly characterized, and comparison studies across the World Health Organization diagnostic categories are lacking. Methods Tumor samples from a total of 102 patient cases within four major subtypes of malignant histiocytic and dendritic cell neoplasms, including 44 follicular dendritic cell sarcomas (FDCSs), 41 histiocytic sarcomas (HSs), 7 interdigitating dendritic cell sarcomas (IDCSs), and 10 Langerhans cell sarcomas (LCSs), underwent hybridization capture with analysis of up to 406 cancer‐related genes. Results Among the entire cohort of 102 patients, CDKN2A mutations were most frequent across subtypes and made up 32% of cases, followed by TP53 mutations (22%). Mitogen‐activated protein kinase (MAPK) pathway mutations were present and enriched among the malignant histiocytosis (M) group (HS, IDCS, and LCS) but absent in FDCS (72% vs. 0%; p < .0001). In contrast, NF‐κB pathway mutations were frequent in FDCSs but rare in M group histiocytoses (61% vs. 12%; p < .0001). Tumor mutational burden was significantly higher in M group histiocytoses as compared with FDCSs (median 4.0/Mb vs. 2.4/Mb; p = .012). We also describe a pediatric patient with recurrent secondary histiocytic sarcoma treated with targeted therapy and interrogated by molecular analysis to identify mechanisms of therapeutic resistance. Conclusion A total of 42 patient tumors (41%) harbored pathogenic mutations that were potentially targetable by approved and/or investigative therapies. Our findings highlight the potential value of molecular testing to enable precise tumor classification, identify candidate oncogenic drivers, and define personalized therapeutic options for patients with these aggressive tumors. Implications for Practice This study presents comprehensive genomic profiling results on 102 patient cases within four major subtypes of malignant histiocytic and dendritic cell neoplasms, including 44 follicular dendritic cell sarcomas (FDCSs), 41 histiocytic sarcomas (HSs), 7 interdigitating dendritic cell sarcomas (IDCSs), and 10 Langerhans cell sarcomas (LCSs). MAPK pathway mutations were present and enriched among the malignant histiocytosis (M) group (HS, IDCS, and LCS) but absent in FDCSs. In contrast, NF‐κB pathway mutations were frequent in FDCSs but rare in M group histiocytosis. A total of 42 patient tumors (41%) harbored pathogenic mutations that were potentially targetable by approved and/or investigative therapies. Histiocytic and dendritic cell neoplasms are a diverse group of tumors arising from the monocytic or dendritic cell lineage. This article presents the molecular characteristics of the four major subtypes of malignant histiocytic and dendritic cell neoplasms, focusing on genomic alterations that could represent therapeutic targets.
Collapse
Affiliation(s)
- Lucas R Massoth
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Yin P Hung
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Judith A Ferry
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Robert P Hasserjian
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Valentina Nardi
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - G Petur Nielsen
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sam Sadigh
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Vinayak Venkataraman
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Martin Selig
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alison M Friedmann
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wesley Samore
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Riza Milante
- Department of Dermatology, Jose R. Reyes Memorial Medical Center, Manila, Philippines
| | - Joseph Giessinger
- A.T. Still University School of Osteopathic Medicine, Mesa, Arizona, USA
| | - Kathleen Foley-Peres
- Department of Biology, Bristol Community College, Fall River, Massachusetts, USA
| | - Chelsea Marcus
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
| | - Eric Severson
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
| | - Daniel Duncan
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA
| | | | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA.,Department of Pathology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Vikram Desphande
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shakti H Ramkissoon
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA.,Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Abner Louissaint
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lawrence R Zukerberg
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Erik A Williams
- Foundation Medicine, Inc., Cambridge, Massachusetts, USA.,Department of Pathology, Department of Dermatology, UCSF Dermatopathology Service, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
58
|
Chakraborty R, Abdel-Wahab O, Durham BH. MAP-Kinase-Driven Hematopoietic Neoplasms: A Decade of Progress in the Molecular Age. Cold Spring Harb Perspect Med 2021; 11:a034892. [PMID: 32601132 PMCID: PMC7770072 DOI: 10.1101/cshperspect.a034892] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Mutations in members of the mitogen-activated protein kinase (MAPK) pathway are extensively studied in epithelial malignancies, with BRAF mutations being one of the most common alterations activating this pathway. However, BRAF mutations are overall quite rare in hematological malignancies. Studies over the past decade have identified high-frequency BRAF V600E, MAP2K1, and other kinase alterations in two groups of MAPK-driven hematopoietic neoplasms: hairy cell leukemia (HCL) and the systemic histiocytoses. Despite HCL and histiocytoses sharing common molecular alterations, these are phenotypically distinct malignancies that differ in respect to clinical presentation and suspected cell of origin. The purpose of this review is to highlight the molecular advancements over the last decade in the histiocytic neoplasms and HCL and discuss the impact these insights have had on our understanding of the molecular pathophysiology, cellular origins, and therapy of these enigmatic diseases as well as perspectives for future research directions.
Collapse
Affiliation(s)
- Rikhia Chakraborty
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, Texas 77030, USA
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Benjamin H Durham
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
59
|
Bigenwald C, Le Berichel J, Wilk CM, Chakraborty R, Chen ST, Tabachnikova A, Mancusi R, Abhyankar H, Casanova-Acebes M, Laface I, Akturk G, Jobson J, Karoulia Z, Martin JC, Grout J, Rafiei A, Lin H, Manz MG, Baccarini A, Poulikakos PI, Brown BD, Gnjatic S, Lujambio A, McClain KL, Picarsic J, Allen CE, Merad M. BRAF V600E-induced senescence drives Langerhans cell histiocytosis pathophysiology. Nat Med 2021; 27:851-861. [PMID: 33958797 PMCID: PMC9295868 DOI: 10.1038/s41591-021-01304-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
Langerhans cell histiocytosis (LCH) is a potentially fatal condition characterized by granulomatous lesions with characteristic clonal mononuclear phagocytes (MNPs) harboring activating somatic mutations in mitogen-activated protein kinase (MAPK) pathway genes, most notably BRAFV600E. We recently discovered that the BRAFV600E mutation can also affect multipotent hematopoietic progenitor cells (HPCs) in multisystem LCH disease. How the BRAFV600E mutation in HPCs leads to LCH is not known. Here we show that enforced expression of the BRAFV600E mutation in early mouse and human multipotent HPCs induced a senescence program that led to HPC growth arrest, apoptosis resistance and a senescence-associated secretory phenotype (SASP). SASP, in turn, promoted HPC skewing toward the MNP lineage, leading to the accumulation of senescent MNPs in tissue and the formation of LCH lesions. Accordingly, elimination of senescent cells using INK-ATTAC transgenic mice, as well as pharmacologic blockade of SASP, improved LCH disease in mice. These results identify senescent cells as a new target for the treatment of LCH.
Collapse
Affiliation(s)
- Camille Bigenwald
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Le Berichel
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Matthias Wilk
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rikhia Chakraborty
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Steven T Chen
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra Tabachnikova
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebecca Mancusi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harshal Abhyankar
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Maria Casanova-Acebes
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilaria Laface
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guray Akturk
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jenielle Jobson
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zoi Karoulia
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jerome C Martin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John Grout
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anahita Rafiei
- Department of Medical Oncology and Hematology, University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Howard Lin
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Alessia Baccarini
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Poulikos I Poulikakos
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amaia Lujambio
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenneth L McClain
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer Picarsic
- Department of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
60
|
Suh JK, Kang S, Kim H, Im HJ, Koh KN. Recent advances in the understanding of the molecular pathogenesis and targeted therapy options in Langerhans cell histiocytosis. Blood Res 2021; 56:S65-S69. [PMID: 33935037 PMCID: PMC8093998 DOI: 10.5045/br.2021.2021013] [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: 01/21/2021] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is the most common histiocytic disorder caused by the clonal expansion of myeloid precursors that differentiate into CD1a+/CD207+ cells in the lesion. Advances in genomic sequencing techniques have improved our understanding of the pathophysiology of LCH. Activation of the mitogen-activated protein kinase (MAPK) pathway is a key molecular mechanism involved in the development of LCH. Recurrent BRAF mutations and MAP2K1 mutations are the major molecular alterations involved in the activation of the MAPK pathway. Recent studies have supported the “misguided myeloid differentiation model” of LCH, where the extent of disease is defined by the differentiation stage of the cell in which the activating somatic MAPK mutation occurs, suggesting LCH. Several studies have advocated the efficacy of targeted therapy using BRAF inhibitors with a high response rate, especially in patients with high-risk or refractory LCH. However, the optimal treatment scheme for children remains unclear. This review outlines recent advances in LCH, focusing on understanding the molecular pathophysiology, emerging targeted therapy options, and their clinical implications.
Collapse
Affiliation(s)
- Jin Kyung Suh
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Sunghan Kang
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyery Kim
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Ho Joon Im
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung-Nam Koh
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
61
|
Wang CJ, Cui L, Ma HH, Wang D, Zhang L, Lian HY, Li WJ, Zhang Q, Wang TY, Li ZG, Zhang R. BRAF V600E Mutation in Cell-Free DNA, Rather than in Lesion Tissues, at Diagnosis Is An Independent Prognostic Factor in Children with Langerhans Cell Histiocytosis. Mol Cancer Ther 2021; 20:1316-1323. [PMID: 33879554 DOI: 10.1158/1535-7163.mct-20-1075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/17/2021] [Accepted: 04/14/2021] [Indexed: 11/16/2022]
Abstract
The aim of this study was to investigate the prognostic significance of BRAFV600E in cell-free (cf) DNA (cfBRAFV600E) and lesion tissues (ltBRAFV600E) in pediatric Langerhans cell histiocytosis (LCH). This study included a total of 140 patients with successfully detected cfBRAFV600E and ltBRAFV600E at diagnosis. Treatment response at week 6 was correlated with both cfBRAFV600E and ltBRAFV600E Moreover, the patients with positive cfBRAFV600E had a much lower 3-year progression-free survival (PFS) rate and a higher progression/reactivation rate than those with negative cfBRAFV600E (47.1% ± 7.6% vs. 78.4% ± 5.1%, P < 0.0001; 44.6% vs. 19.0%, P = 0.001, respectively). However, no significant difference was found in the 3-year PFS rate or progression/reactivation rate between patients with positive and negative ltBRAFV600E (P = 0.348 and 0.596, respectively). In addition, after patients were divided into group A (both cfBRAFV600E and ltBRAFV600E positive, n = 56), group B (ltBRAFV600E positive and cfBRAFV600E negative, n = 28), and group C (both cfBRAFV600E and ltBRAFV600E negative, n = 56), there was a significant difference in the 3-year PFS rate and progression/reactivation rate among the three groups (47.1% ± 7.6%, 92.9% ± 6.1%, and 72.2% ± 6.1%, P < 0.001; 44.6%, 3.6%, and 26.8%, P < 0.001, respectively). In the multivariate analysis, cfBRAFV600E and age at diagnosis remained independent prognostic factors for 3-year PFS in childhood LCH. Therefore, cfBRAFV600E was more closely associated with important clinical characteristics, treatment response at week 6, and prognosis than ltBRAFV600E.
Collapse
Affiliation(s)
- Chan-Juan Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lei Cui
- Hematologic Disease Laboratory, Hematology Center, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hong-Hao Ma
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Dong Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Li Zhang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hong-Yun Lian
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei-Jing Li
- Hematologic Disease Laboratory, Hematology Center, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qing Zhang
- Hematologic Disease Laboratory, Hematology Center, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Tian-You Wang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Zhi-Gang Li
- Hematologic Disease Laboratory, Hematology Center, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| | - Rui Zhang
- Hematology Center, Beijing Key Laboratory of Pediatric Hematology Oncology; National Key Discipline of Pediatrics (Capital Medical University); Key Laboratory of Major Diseases in Children, Ministry of Education; Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| |
Collapse
|
62
|
Sengal A, Velazquez J, Hahne M, Burke TM, Abhyankar H, Reyes R, Olea W, Scull B, Eckstein OS, Bigenwald C, Bollard CM, Yu W, Merad M, McClain KL, Allen CE, Chakraborty R. Overcoming T-cell exhaustion in LCH: PD-1 blockade and targeted MAPK inhibition are synergistic in a mouse model of LCH. Blood 2021; 137:1777-1791. [PMID: 33075814 PMCID: PMC8020265 DOI: 10.1182/blood.2020005867] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasia characterized by granulomatous lesions containing pathological CD207+ dendritic cells (DCs) with persistent MAPK pathway activation. Standard-of-care chemotherapies are inadequate for most patients with multisystem disease, and optimal strategies for relapsed and refractory disease are not defined. The mechanisms underlying development of inflammation in LCH lesions, the role of inflammation in pathogenesis, and the potential for immunotherapy are unknown. Analysis of the immune infiltrate in LCH lesions identified the most prominent immune cells as T lymphocytes. Both CD8+ and CD4+ T cells exhibited "exhausted" phenotypes with high expression of the immune checkpoint receptors. LCH DCs showed robust expression of ligands to checkpoint receptors. Intralesional CD8+ T cells showed blunted expression of Tc1/Tc2 cytokines and impaired effector function. In contrast, intralesional regulatory T cells demonstrated intact suppressive activity. Treatment of BRAFV600ECD11c LCH mice with anti-PD-1 or MAPK inhibitor reduced lesion size, but with distinct responses. Whereas MAPK inhibitor treatment resulted in reduction of the myeloid compartment, anti-PD-1 treatment was associated with reduction in the lymphoid compartment. Notably, combined treatment with MAPK inhibitor and anti-PD-1 significantly decreased both CD8+ T cells and myeloid LCH cells in a synergistic fashion. These results are consistent with a model that MAPK hyperactivation in myeloid LCH cells drives recruitment of functionally exhausted T cells within the LCH microenvironment, and they highlight combined MAPK and checkpoint inhibition as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Amel Sengal
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and
| | - Jessica Velazquez
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and
| | - Meryl Hahne
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Thomas M Burke
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and
- Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | - Harshal Abhyankar
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Robert Reyes
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Walter Olea
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Brooks Scull
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Olive S Eckstein
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Camille Bigenwald
- Department of Oncological Sciences, Tisch Cancer Institute, and
- Department of Dermatology, Icahn School of Medicine, New York, NY
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Medical Center, Washington, DC
- Department of Pediatrics and
- Department of Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC; and
| | - Wendong Yu
- Department of Pathology, Baylor College of Medicine, Houston, TX
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute, and
- Department of Dermatology, Icahn School of Medicine, New York, NY
| | - Kenneth L McClain
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and
| | - Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and
- Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | - Rikhia Chakraborty
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and
| |
Collapse
|
63
|
Bone marrow-derived myeloid progenitors as driver mutation carriers in high- and low-risk Langerhans cell histiocytosis. Blood 2021; 136:2188-2199. [PMID: 32750121 DOI: 10.1182/blood.2020005209] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is a myeloid neoplasia, driven by sporadic activating mutations in the MAPK pathway. The misguided myeloid dendritic cell (DC) model proposes that high-risk, multisystem, risk-organ-positive (MS-RO+) LCH results from driver mutation in a bone marrow (BM)-resident multipotent hematopoietic progenitor, while low-risk, MS-RO- and single-system LCH would result from driver mutation in a circulating or tissue-resident, DC-committed precursor. We have examined the CD34+c-Kit+Flt3+ myeloid progenitor population as potential mutation carrier in all LCH disease manifestations. This population contains oligopotent progenitors of monocytes (Mo's)/macrophages (MΦs), osteoclasts (OCs), and DCs. CD34+c-Kit+Flt3+ cells from BM of MS-RO+ LCH patients produced Langerhans cell (LC)-like cells in vitro. Both LC-like and DC offspring from this progenitor carried the BRAF mutation, confirming their common origin. In both high- and low-risk LCH patients, CD34+c-Kit+Flt3+ progenitor frequency in blood was higher than in healthy donors. In one MS-RO+ LCH patient, CD34+c-Kit+Flt3+ cell frequency in blood and its BRAF-mutated offspring reported response to chemotherapy. CD34+c-Kit+Flt3+ progenitors from blood of both high- and low-risk LCH patients gave rise to DCs and LC-like cells in vitro, but the driver mutation was not easily detectable, likely due to low frequency of mutated progenitors. Mutant BRAF alleles were found in Mo's /MΦs, DCs, LC-like cells, and/or OC-like cells in lesions and/or Mo and DCs in blood of multiple low-risk patients. We therefore hypothesize that in both high- and low-risk LCH, the driver mutation is present in a BM-resident myeloid progenitor that can be mobilized to the blood.
Collapse
|
64
|
Indeterminate Dendritic Cell Tumor With Persistent Complete Metabolic Response to BRAF/MEK Inhibition. Hemasphere 2020; 5:e511. [PMID: 33324955 PMCID: PMC7732267 DOI: 10.1097/hs9.0000000000000511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 10/21/2020] [Indexed: 11/26/2022] Open
|
65
|
Affiliation(s)
- Nitya Gulati
- Texas Children's Cancer Center and Hematology Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
66
|
Abstract
Langerhans cell histiocytosis (LCH) is caused by clonal expansion of myeloid precursors that differentiate into CD1a+/CD207+ cells in lesions that leads to a spectrum of organ involvement and dysfunction. The pathogenic cells are defined by constitutive activation of the MAPK signaling pathway. Treatment of LCH is risk-adapted: patients with single lesions may respond well to local treatment, whereas patients with multisystem disease require systemic therapy. Although survival rates for patients without organ dysfunction is excellent, mortality rates for patients with organ dysfunction may reach 20%. Despite progress made in the treatment of LCH, disease reactivation rates remain above 30%, and standard second-line treatment is yet to be established. Treatment failure is associated with increased risks for death and long-term morbidity, including LCH-associated neurodegeneration. Early case series report promising clinical responses in patients with relapsed and refractory LCH treated with BRAF or MEK inhibitors, although potential for this strategy to achieve cure remains uncertain.
Collapse
|
67
|
Jouenne F, Sadoux A, Lorillon G, Louveau B, Bugnet E, Meignin V, Mourah S, Tazi A. Custom pyrosequencing assay to detect short BRAF deletions in Langerhans cell histiocytic lesions. J Clin Pathol 2020; 74:533-536. [PMID: 32873703 DOI: 10.1136/jclinpath-2020-206974] [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: 07/23/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 11/04/2022]
Abstract
Langerhans cell histiocytosis (LCH) is a rare inflammatory myeloid neoplastic disease driven by activating mutations in the mitogen-activating protein kinase signalling pathway, including the BRAF V600E mutation and BRAF deletions (BRAFdel). Next-generation sequencing and whole exome sequencing (WES) are valuable and powerful approaches for BRAFdel identification, but these techniques are costly and time consuming. Pyrosequencing is an alternative method that has the potential to rapidly and reliably identify gene deletions. We developed a custom pyrosequencing assay to detect the exon-12 BRAFdel in 18 biopsies from adult patients with LCH, which were all genotyped in parallel using Sanger sequencing and WES. A BRAFdel was detected in 7/18 (39%), 6/18 (33%) and 3/18 (17%) LCH lesions using WES, pyrosequencing and Sanger, respectively, with good concordance between the WES and pyrosequencing results (Kappa-coefficient=0.88). Therefore, our pyrosequencing assay is reliable and useful for detecting BRAFdel, particularly in BRAF V600E-negative LCH lesions, for which targeted treatment is indicated.
Collapse
Affiliation(s)
- Fanélie Jouenne
- Pharmacogenomics Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France.,INSERM U976, Université de Paris, Paris, Île-de-France, France
| | - Aurélie Sadoux
- Pharmacogenomics Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France.,INSERM U976, INSERM, Paris, Île-de-France, France
| | - Gwenaël Lorillon
- National Reference Centre for Histiocytoses, Pulmonology Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| | - Baptiste Louveau
- Pharmacogenomics Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France.,INSERM U976, INSERM, Paris, Île-de-France, France
| | - Emmanuelle Bugnet
- National Reference Centre for Histiocytoses, Pulmonology Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| | - Veronique Meignin
- Pathology Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| | - Samia Mourah
- Pharmacogenomics Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France.,INSERM U976, Université de Paris, Paris, Île-de-France, France
| | - Abdellatif Tazi
- INSERM U976, Université de Paris, Paris, Île-de-France, France .,National Reference Centre for Histiocytoses, Pulmonology Department, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, Île-de-France, France
| |
Collapse
|
68
|
Li N, Cui L, Ma H, Gong Z, Lian H, Wang C, Zhang Q, Zhao X, Chen X, Tian Y, Yang Y, Wei A, Zhang R, Li Z, Wang T. Osteopontin is highly secreted in the cerebrospinal fluid of patient with posterior pituitary involvement in Langerhans cell histiocytosis. Int J Lab Hematol 2020; 42:788-795. [PMID: 32845092 DOI: 10.1111/ijlh.13304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 07/06/2020] [Accepted: 07/13/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Langerhans cell histiocytosis (LCH) is a rare disease caused by clonal proliferation of CD1a+ CD207+ cells. Distinguishing pituitary involvement was essential in stratification and treatment of patients with LCH. The diagnosis of pituitary involvement is mainly dependent on hormone abnormalities in the anterior pituitary and magnetic resonance imaging (MRI) scanning in posterior pituitary. Diabetes insipidus (DI) is a serious sequelae and often occurred with pituitary involvement. It is reported that osteopontin (OPN) is highly secreted in the cerebrospinal fluid (CSF) of patients with neurodegenerative diseases in LCH (LCH-ND). However, patients with posterior pituitary involvement account for a larger portion in our hospital. Whether the OPN level could be an auxiliary diagnostic marker for the posterior pituitary involvement or not is still unknown. METHODS In our study, we collected CSF samples of 57 children with LCH. The secreted OPN (sOPN) levels in CSF were measured through enzyme-linked immunosorbent assay (ELISA). RESULTS After the retrospective analysis of 57 patients with LCH, we found that the sOPN levels in CSF of children with posterior pituitary involvement were significantly higher than that of other groups. After the Pearson Chi-Square test, Fisher's exact test and ROC analysis, we found that the sOPN levels were significantly correlated with posterior pituitary involvement. The cut-off value is 214.14 ng/mL. CONCLUSION The sOPN levels were elevated in CSF of LCH children with posterior pituitary involvement. Analysis of the sOPN level may provide more accurate auxiliary diagnostic techniques for the clinic.
Collapse
Affiliation(s)
- Na Li
- Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Lei Cui
- Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Honghao Ma
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Zhihua Gong
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Taiyuan, China
| | - Hongyun Lian
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chanjuan Wang
- Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qing Zhang
- Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoxi Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xihua Chen
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yu Tian
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ying Yang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ang Wei
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Rui Zhang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University & Capital Medical University, Beijing Children's Hospital, Beijing, China
| | - Zhigang Li
- Laboratory of Hematologic Diseases, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Tianyou Wang
- Beijing Key Laboratory of Pediatric Hematology Oncology, National Key Discipline of Pediatrics, Capital Medical University, Key Laboratory of Major Diseases in Children, Ministry of Education, Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
69
|
Botton T, Talevich E, Mishra VK, Zhang T, Shain AH, Berquet C, Gagnon A, Judson RL, Ballotti R, Ribas A, Herlyn M, Rocchi S, Brown KM, Hayward NK, Yeh I, Bastian BC. Genetic Heterogeneity of BRAF Fusion Kinases in Melanoma Affects Drug Responses. Cell Rep 2020; 29:573-588.e7. [PMID: 31618628 DOI: 10.1016/j.celrep.2019.09.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 07/26/2019] [Accepted: 09/04/2019] [Indexed: 12/15/2022] Open
Abstract
BRAF fusions are detected in numerous neoplasms, but their clinical management remains unresolved. We identified six melanoma lines harboring BRAF fusions representative of the clinical cases reported in the literature. Their unexpected heterogeneous responses to RAF and MEK inhibitors could be categorized upon specific features of the fusion kinases. Higher expression level correlated with resistance, and fusion partners containing a dimerization domain promoted paradoxical activation of the mitogen-activated protein kinase (MAPK) pathway and hyperproliferation in response to first- and second-generation RAF inhibitors. By contrast, next-generation αC-IN/DFG-OUT RAF inhibitors blunted paradoxical activation across all lines and had their therapeutic efficacy further increased in vitro and in vivo by combination with MEK inhibitors, opening perspectives in the clinical management of tumors harboring BRAF fusions.
Collapse
Affiliation(s)
- Thomas Botton
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA.
| | - Eric Talevich
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Vivek Kumar Mishra
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Tongwu Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MA 20892, USA
| | - A Hunter Shain
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Céline Berquet
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Alexander Gagnon
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Robert L Judson
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Robert Ballotti
- U1065, Institut National de la Santé et de la Recherche Médicale, Centre Méditerranéen de Médecine Moléculaire, Université Côte d'Azur, 06200 Nice, France
| | - Antoni Ribas
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program and Melanoma Research Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Stéphane Rocchi
- U1065, Institut National de la Santé et de la Recherche Médicale, Centre Méditerranéen de Médecine Moléculaire, Université Côte d'Azur, 06200 Nice, France
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MA 20892, USA
| | - Nicholas K Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Iwei Yeh
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Boris C Bastian
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94115, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94115, USA.
| |
Collapse
|
70
|
Zhang B, Chen Y, Dai P, Yu H, Ma J, Chen C, Zhang Y, Guan Y, Chen R, Liu T, Wang J, Yang L, Yi X, Xia X, Ma H. Oncogenic mutations within the β3-αC loop of EGFR/ERBB2/BRAF/MAP2K1 predict response to therapies. Mol Genet Genomic Med 2020; 8:e1395. [PMID: 32757330 PMCID: PMC7549570 DOI: 10.1002/mgg3.1395] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/31/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Background β3‐αC loop is a highly conserved structural domain across oncogene families, which is a switch for kinase activity. There have been numerous researches on mutations within β3‐αC loop in EGFR, but relatively less in ERBB2, BRAF, and MAP2K1. In addition, previous studies mainly focus on β3‐αC deletion in EGFR, which is the most common type affecting kinase activity and driving lung cancer. Other mutation types are not well studied. Methods Here we analyzed the profile of β3‐αC loop mutations in a total of 10,000 tumor biopsy and/or ctDNA patient samples using hybridization capture‐based next‐generation sequencing. Results We identified 1616 mutations within β3‐αC loop in this cohort. Most mutations were located in EGFR, with less percentage in ERBB2, BRAF, and MAP2K1. EGFR β3‐αC deletions occurred at a high percentage of 96.7% and were all drug‐relevant. We also detected rare EGFR β3‐αC insertions and point mutations, most of which were related to EGFR TKIs resistance. ERBB2 β3‐αC deletions were only found in breast cancers and sensitive to EGFR/ERBB2 inhibitor. Moreover, BRAF and MAP2K1 mutations within β3‐αC loop also demonstrated drugs relevance. Conclusion Our study showed that oncogenic mutations within the β3‐αC loop of ERBB2, MAP2K1, and BRAF are analogous to that of EGFR, which have profound effect on drug response. Understanding the mutation profile within the β3‐αC loop is critical for targeted therapies.
Collapse
Affiliation(s)
- Biao Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongsheng Chen
- Geneplus-Beijing, Beijing, China.,Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Pingping Dai
- Geneplus-Beijing, Beijing, China.,Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Haoda Yu
- Department of Respiratory Medicine, Wuxi People's Hospital, Wuxi, China
| | | | - Chen Chen
- Geneplus-Beijing, Beijing, China.,Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhang
- Geneplus-Beijing, Beijing, China.,Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Yanfang Guan
- Geneplus-Beijing, Beijing, China.,Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | | | - Tao Liu
- Geneplus-Beijing, Beijing, China.,Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Jiayin Wang
- Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | | | - Xin Yi
- Geneplus-Beijing, Beijing, China
| | | | - Haitao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
71
|
Hayase T, Saito S, Shioda Y, Imamura T, Watanabe K, Ohki K, Yoshioka T, Oh Y, Kawahara Y, Niijima H, Imashuku S, Morimoto A. Analysis of the BRAF and MAP2K1 mutations in patients with Langerhans cell histiocytosis in Japan. Int J Hematol 2020; 112:560-567. [PMID: 32654047 DOI: 10.1007/s12185-020-02940-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/11/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
In Langerhans cell histiocytosis (LCH), somatic gene mutations in the mitogen-activated protein kinase pathway have been identified in more than 80% of cases in Western countries, in which mutually exclusive BRAF and MAP2K1 mutations are involved. Among them, BRAF V600E mutation is the major contributor (50-60%). In 59 patients (50 children and nine adults) with LCH (not including pulmonary LCH) in Japan, we first screened for BRAF V600E in all patients followed by target sequencing for other gene mutations in 17 of BRAF V600E-negative patients. As a result, BRAF V600E mutation was detected in 27/59 (46%) patients. We also identified BRAF mutations other than V600E in five and MAP2K1 mutations in nine patients. Thus, gene mutations in BRAF or MAP2K1 were identified in 41/44 (93%) of the fully tested patients. Regarding the correlation of clinical features and genotype in pediatric patients, we found that BRAF V600E mutation status was not correlated with sex, age at diagnosis, disease extent, response to first-line therapy, relapse, or CNS-related sequelae. Interestingly, MAP2K1 exon 2 in-frame deletion was related to the risk organ involvement; however, further studies are required to clarify the impact of these gene mutations on the clinical features of patients with LCH.
Collapse
Affiliation(s)
- Tomomi Hayase
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shiori Saito
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yoko Shioda
- Department of Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenichiro Watanabe
- Department of Hematology and Oncology, Shizuoka Children's Hospital, Shizuoka, Japan
| | - Kentaro Ohki
- Department of Pediatric Hematology and Oncology Research, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takako Yoshioka
- Department of Pathology, National Center for Child Health and Development, Tokyo, Japan
| | - Yukiko Oh
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yuta Kawahara
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hitomi Niijima
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shinsaku Imashuku
- Division of Laboratory Medicine, Uji Tokushukai Medical Center, Uji, Japan
| | - Akira Morimoto
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| |
Collapse
|
72
|
The combination of methotrexate and cytosine arabinoside in newly diagnosed adult Langerhans cell histiocytosis: a prospective phase II interventional clinical trial. BMC Cancer 2020; 20:433. [PMID: 32423455 PMCID: PMC7236107 DOI: 10.1186/s12885-020-06872-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 04/15/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Langerhans Cell Histiocytosis (LCH) is a rare disease puzzling both children and adults, however outcome of adult patients is unfavorable. This prospective interventional trial aims to test the efficacy and safety of the combination of methotrexate and cytosine arabinoside in adult LCH patients. METHOD A total of 36 patients enrolled diagnosed with LCH and treated in our center from 1st Jan, 2014 to 30th Jun, 2016. RESULT Nineteen patients underwent the detection of BRAF mutation, with a positive rate of 21.1%. The overall response rate was 100%, only 16.7% achieved complete response. The overall regression rate of osseous lesions was 100%. Regression of central nervous system involvement was also favorable. After a median follow-up of 44 months, the estimated event-free survival was 48.9 months, the overall survival rate was 97.2%. The risk organ involvement showed strong prognostic value, EFS was 34.1 or 54.6 months (p = 0.001) in groups with/without risk organ involvement respectively. Neutropenia and thrombocytopenia were the most common adverse effects. CONCLUSION The regimen of methotrexate and cytosine arabinoside (MA) is effective and safe in treating adult LCH patients, and timely preventions may be considered for the high incidence of hematological adverse effects. TRIAL REGISTRATION Trial No. NCT02389400 on Clinicaltrials.gov, registered on 10th Mar. 2015.
Collapse
|
73
|
Thompson ER, Lim KJC, Kuzich JA, McBean M, Westerman D, Tam CS, Blombery P. Detection of an IGH- BRAF fusion in a patient with BRAF Val600Glu negative hairy cell leukemia. Leuk Lymphoma 2020; 61:2024-2026. [PMID: 32319330 DOI: 10.1080/10428194.2020.1753045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Ella R Thompson
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Kenneth J C Lim
- Department of Haematology, St Vincent's Hospital, Melbourne, Australia
| | - James A Kuzich
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michelle McBean
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - David Westerman
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia
| | - Constantine S Tam
- Department of Haematology, St Vincent's Hospital, Melbourne, Australia.,Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Piers Blombery
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Clinical Haematology, Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
74
|
Louveau B, Jouenne F, Kaguelidou F, Landras A, Goldwirt L, Mourah S. The key role of oncopharmacology in therapeutic management, from common to rare cancers: A literature review. Therapie 2020; 75:183-193. [DOI: 10.1016/j.therap.2020.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/15/2019] [Indexed: 01/18/2023]
|
75
|
Polypoid Atypical Spitz Tumor With a Fibrosclerotic Stroma, CLIP2-BRAF Fusion, and Homozygous Loss of 9p21. Am J Dermatopathol 2020; 42:204-207. [DOI: 10.1097/dad.0000000000001502] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
76
|
Jouenne F, Chevret S, Bugnet E, Clappier E, Lorillon G, Meignin V, Sadoux A, Cohen S, Haziot A, How-Kit A, Kannengiesser C, Lebbé C, Gossot D, Mourah S, Tazi A. Genetic landscape of adult Langerhans cell histiocytosis with lung involvement. Eur Respir J 2020; 55:13993003.01190-2019. [PMID: 31806714 DOI: 10.1183/13993003.01190-2019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022]
Abstract
The clinical significance of the BRAF V600E mutation in adult Langerhans cell histiocytosis (LCH), including pulmonary Langerhans cell histiocytosis (PLCH), is not well understood. Similarly, the spectrum of molecular alterations involved in adult LCH has not been fully delineated. To address these issues, we genotyped a large number of adult LCH biopsies and searched for an association of identified molecular alterations with clinical presentation and disease outcome.Biopsies from 117 adult LCH patients, 83 with PLCH (median age 36.4 years, 56 females, 38 multisystem disease, 79 single system disease, 65 current smokers) were genotyped for the BRAF V600E mutation. In 69 cases, LCH lesions were also genotyped by whole-exome sequencing (WES) or targeted gene panel next-generation sequencing (NGS). Cox models were used to estimate the association of baseline characteristics with the hazard of LCH progression.MAPK pathway alterations were detected in 59 out of 69 cases (86%) (BRAF V600E mutation: 36%, BRAF N486_P490 deletion: 28%, MAP2K1 mutations: 15%, isolated NRAS Q61 mutations: 4%), while KRAS mutations were virtually absent in PLCH lesions. The BRAF V600E mutation was not associated with LCH presentation at diagnosis, including smoking status and lung function, in PLCH patients. BRAF V600E status did not influence the risk of LCH progression over time.Thus, MAPK alterations are present in most lesions from adult LCH patients, particularly in PLCH. Unlike reports in paediatric LCH, BRAF V600E genotyping did not provide additional information on disease outcome. The search for alterations involved in the MAPK pathway, including BRAF deletions, is useful for guiding targeted treatment in selected patients with refractory progressive LCH.
Collapse
Affiliation(s)
- Fanélie Jouenne
- Université de Paris, INSERM U976, Institut de Recherche Saint-Louis, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Laboratoire de Pharmacogénomique, Paris, France
| | - Sylvie Chevret
- Université de Paris, U1153 CRESS, Équipe de Recherche en Biostatistiques et Épidémiologie Clinique (ECSTRRA), Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Service de Biostatistique et Information Médicale, Paris, France
| | - Emmanuelle Bugnet
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Centre National de Référence des Histiocytoses, Service de Pneumologie, Paris, France
| | - Emmanuelle Clappier
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Laboratoire d'Hématologie Biologique, Paris, France.,Université de Paris, INSERM U944, Institut de Recherche Saint-Louis, Paris, France
| | - Gwenaël Lorillon
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Centre National de Référence des Histiocytoses, Service de Pneumologie, Paris, France
| | - Véronique Meignin
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Service de Pathologie, INSERM UMR_S1165, Paris, France
| | - Aurélie Sadoux
- Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Laboratoire de Pharmacogénomique, Paris, France
| | - Shannon Cohen
- INSERM U1160, Institut de Recherche Saint-Louis, Paris, France
| | - Alain Haziot
- INSERM U1160, Institut de Recherche Saint-Louis, Paris, France
| | - Alexandre How-Kit
- Laboratoire de Génomique Fonctionnelle, Fondation Jean Dausset - CEPH, Paris, France
| | - Caroline Kannengiesser
- Université de Paris, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Laboratoire de Génétique, Paris, France
| | - Céleste Lebbé
- Université de Paris, INSERM U976, Institut de Recherche Saint-Louis, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Département de Dermatologie, Paris, France
| | - Dominique Gossot
- Institut du Thorax Curie-Montsouris, Département Thoracique, Institut Mutualiste Montsouris, Paris, France
| | - Samia Mourah
- Université de Paris, INSERM U976, Institut de Recherche Saint-Louis, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Laboratoire de Pharmacogénomique, Paris, France
| | - Abdellatif Tazi
- Université de Paris, INSERM U976, Institut de Recherche Saint-Louis, Paris, France .,Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Centre National de Référence des Histiocytoses, Service de Pneumologie, Paris, France
| |
Collapse
|
77
|
Liu H, Osterburg AR, Flury J, Swank Z, McGraw DW, Gupta N, Wikenheiser-Brokamp KA, Kumar A, Tazi A, Inoue Y, Hirose M, McCormack FX, Borchers MT. MAPK mutations and cigarette smoke promote the pathogenesis of pulmonary Langerhans cell histiocytosis. JCI Insight 2020; 5:132048. [PMID: 31961828 DOI: 10.1172/jci.insight.132048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary Langerhans cell histiocytosis (PLCH) is a rare smoking-related lung disease characterized by dendritic cell (DC) accumulation, bronchiolocentric nodule formation, and cystic lung remodeling. Approximately 50% of patients with PLCH harbor somatic BRAF-V600E mutations in cells of the myeloid/monocyte lineage. However, the rarity of the disease and lack of animal models have impeded the study of PLCH pathogenesis. Here, we establish a cigarette smoke-exposed (CS-exposed) BRAF-V600E-mutant mouse model that recapitulates many hallmark characteristics of PLCH. We show that CD11c-targeted expression of BRAF-V600E increases DC responsiveness to stimuli, including the chemokine CCL20, and that mutant cell accumulation in the lungs of CS-exposed mice is due to both increased cellular viability and enhanced recruitment. Moreover, we report that the chemokine CCL7 is secreted from DCs and human peripheral blood monocytes in a BRAF-V600E-dependent manner, suggesting a possible mechanism for recruitment of cells known to dominate PLCH lesions. Inflammatory lesions and airspace dilation in BRAF-V600E mice in response to CS are attenuated by transitioning animals to filtered air and treatment with a BRAF-V600E inhibitor, PLX4720. Collectively, this model provides mechanistic insights into the role of myelomonocytic cells and the BRAF-V600E mutation and CS exposure in PLCH pathogenesis and provides a platform to develop biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Huan Liu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andrew R Osterburg
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer Flury
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Zulma Swank
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Dennis W McGraw
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Nishant Gupta
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pathology and Laboratory Medicine and.,Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ashish Kumar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Abdellatif Tazi
- INSERM UMR-S 976, University Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Yoshikazu Inoue
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Masaki Hirose
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Michael T Borchers
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
78
|
Lim KPH, Milne P, Poidinger M, Duan K, Lin H, McGovern N, Abhyankar H, Zinn D, Burke TM, Eckstein OS, Chakraborty R, Sengal A, Scull B, Newell E, Merad M, McClain KL, Man TK, Ginhoux F, Collin M, Allen CE. Circulating CD1c+ myeloid dendritic cells are potential precursors to LCH lesion CD1a+CD207+ cells. Blood Adv 2020; 4:87-99. [PMID: 31899802 PMCID: PMC6960472 DOI: 10.1182/bloodadvances.2019000488] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/24/2019] [Indexed: 12/17/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is a myeloproliferative disorder that is characterized by the inflammatory lesions with pathogenic CD1a+CD207+ dendritic cells (DCs). BRAFV600E and other somatic activating MAPK gene mutations have been identified in differentiating bone marrow and blood myeloid cells, but the origin of the LCH lesion CD1a+CD207+ DCs and mechanisms of lesion formation remain incompletely defined. To identify candidate LCH CD1a+CD207+ DC precursor populations, gene-expression profiles of LCH lesion CD1a+CD207+ DCs were first compared with established gene signatures from human myeloid cell subpopulations. Interestingly, the CD1c+ myeloid DC (mDC) gene signature was most enriched in the LCH CD1a+CD207+ DC transcriptome. Additionally, the BRAFV600E allele was not only localized to CD1a+CD207- DCs and CD1a+CD207+ DCs, but it was also identified in CD1c+ mDCs in LCH lesions. Transcriptomes of CD1a+CD207- DCs were nearly indistinguishable from CD1a+CD207+ DCs (both CD1a+CD207low and CD1a+CD207high subpopulations). Transcription profiles of LCH lesion CD1a+CD207+ DCs and peripheral blood CD1c+ mDCs from healthy donors were compared to identify potential LCH DC-specific biomarkers: HLA-DQB2 expression was significantly increased in LCH lesion CD1a+CD207+ DCs compared with circulating CD1c+ mDCs from healthy donors. HLA-DQB2 antigen was identified on LCH lesion CD1a+CD207- DCs and CD1a+CD207+ DCs as well as on CD1c+(CD1a+CD207-) mDCs, but it was not identified in any other lesion myeloid subpopulations. HLA-DQB2 expression was specific to peripheral blood of patients with BRAFV600E+ peripheral blood mononuclear cells, and HLA-DQB2+CD1c+ blood cells were highly enriched for the BRAFV600E in these patients. These data support a model in which blood CD1c+HLA-DQB2+ mDCs with activated ERK migrate to lesion sites where they differentiate into pathogenic CD1a+CD207+ DCs.
Collapse
Affiliation(s)
- Karen Phaik Har Lim
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
- Graduate Program in Translational Biology and Molecular Medicine, College of Medicine, Baylor University, Houston, TX
| | - Paul Milne
- Human Dendritic Cell Laboratory, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kaibo Duan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Howard Lin
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Harshal Abhyankar
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Daniel Zinn
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Thomas M Burke
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
- Graduate Program in Translational Biology and Molecular Medicine, College of Medicine, Baylor University, Houston, TX
- Medical Scientist Training Program, College of Medicine, Baylor University, Houston, TX; and
| | - Olive S Eckstein
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Rikhia Chakraborty
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Amel Sengal
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Brooks Scull
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Evan Newell
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kenneth L McClain
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Tsz-Kwong Man
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Matthew Collin
- Human Dendritic Cell Laboratory, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, and
- Graduate Program in Translational Biology and Molecular Medicine, College of Medicine, Baylor University, Houston, TX
| |
Collapse
|
79
|
Kemps PG, Zondag TC, Steenwijk EC, Andriessen Q, Borst J, Vloemans S, Roelen DL, Voortman LM, Verdijk RM, van Noesel CJM, Cleven AHG, Hawkins C, Lang V, de Ru AH, Janssen GMC, Haasnoot GW, Franken KLMC, van Eijk R, Solleveld-Westerink N, van Wezel T, Egeler RM, Beishuizen A, van Laar JAM, Abla O, van den Bos C, van Veelen PA, van Halteren AGS. Apparent Lack of BRAF V600E Derived HLA Class I Presented Neoantigens Hampers Neoplastic Cell Targeting by CD8 + T Cells in Langerhans Cell Histiocytosis. Front Immunol 2020; 10:3045. [PMID: 31998317 PMCID: PMC6967030 DOI: 10.3389/fimmu.2019.03045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Langerhans Cell Histiocytosis (LCH) is a neoplastic disorder of hematopoietic origin characterized by inflammatory lesions containing clonal histiocytes (LCH-cells) intermixed with various immune cells, including T cells. In 50-60% of LCH-patients, the somatic BRAF V600E driver mutation, which is common in many cancers, is detected in these LCH-cells in an otherwise quiet genomic landscape. Non-synonymous mutations like BRAF V600E can be a source of neoantigens capable of eliciting effective antitumor CD8+ T cell responses. This requires neopeptides to be stably presented by Human Leukocyte Antigen (HLA) class I molecules and sufficient numbers of CD8+ T cells at tumor sites. Here, we demonstrate substantial heterogeneity in CD8+ T cell density in n = 101 LCH-lesions, with BRAF V600E mutated lesions displaying significantly lower CD8+ T cell:CD1a+ LCH-cell ratios (p = 0.01) than BRAF wildtype lesions. Because LCH-lesional CD8+ T cell density had no significant impact on event-free survival, we investigated whether the intracellularly expressed BRAF V600E protein is degraded into neopeptides that are naturally processed and presented by cell surface HLA class I molecules. Epitope prediction tools revealed a single HLA class I binding BRAF V600E derived neopeptide (KIGDFGLATEK), which indeed displayed strong to intermediate binding capacity to HLA-A*03:01 and HLA-A*11:01 in an in vitro peptide-HLA binding assay. Mass spectrometry-based targeted peptidomics was used to investigate the presence of this neopeptide in HLA class I presented peptides isolated from several BRAF V600E expressing cell lines with various HLA genotypes. While the HLA-A*02:01 binding BRAF wildtype peptide KIGDFGLATV was traced in peptides isolated from all five cell lines expressing this HLA subtype, KIGDFGLATEK was not detected in the HLA class I peptidomes of two distinct BRAF V600E transduced cell lines with confirmed expression of HLA-A*03:01 or HLA-A*11:01. These data indicate that the in silico predicted HLA class I binding and proteasome-generated neopeptides derived from the BRAF V600E protein are not presented by HLA class I molecules. Given that the BRAF V600E mutation is highly prevalent in chemotherapy refractory LCH-patients who may qualify for immunotherapy, this study therefore questions the efficacy of immune checkpoint inhibitor therapy in LCH.
Collapse
Affiliation(s)
- Paul G Kemps
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Timo C Zondag
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eline C Steenwijk
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Quirine Andriessen
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Jelske Borst
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - Sandra Vloemans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Dave L Roelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Lenard M Voortman
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Robert M Verdijk
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Carel J M van Noesel
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Cynthia Hawkins
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Veronica Lang
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Arnoud H de Ru
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - George M C Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Geert W Haasnoot
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Kees L M C Franken
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Ronald van Eijk
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Tom van Wezel
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - R Maarten Egeler
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands.,Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Auke Beishuizen
- Department of Pediatric Oncology, Sophia Children's Hospital, Erasmus University Medical Center, Rotterdam, Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Jan A M van Laar
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Oussama Abla
- Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Cor van den Bos
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Pediatric Oncology, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Peter A van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, Netherlands
| | - Astrid G S van Halteren
- Immunology Laboratory Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
80
|
Lian T, Li C, Wang H. Trametinib in the treatment of multiple malignancies harboring MEK1 mutations. Cancer Treat Rev 2019; 81:101907. [PMID: 31715422 DOI: 10.1016/j.ctrv.2019.101907] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/26/2019] [Accepted: 10/05/2019] [Indexed: 01/02/2023]
Abstract
The aberrant activation of RAS-derived mitogen-activated protein kinase (MAPK) signaling pathway plays a prominent role in tumorigenesis of an array of malignancies. The reasons are usually the upstream activated mutations including mitogen-activated protein kinase kinase 1/2 (MEK1/2). As oncogenic mutations, MEK1 mutations have been observed in a variety of malignancies including melanoma, histiocytic neoplasms, colorectal cancer and lung cancer. Presently, the use of trametinib, a highly selective MEK1/2 inhibitor, was limited to BRAF mutations, according to the approvals of FDA. Therefore, we consider that this is a question worth studying that whether malignancies with MEK1 mutations are sensitive to the treatment of trametinib. This review discussed the function of MEK1 mutations, retrieved the frequency and distribution of MEK1 mutations in various malignancies, and reviewed the basic experiments and clinical case reports on trametinib in the treatment of cell lines or patients with MEK1 mutations. Most studies have demonstrated that trametinib was effective to cells or tumor patients harboring MEK1 mutations, which suggest that the MEK1 mutations might be potential indications of trametinib therapy. In addition, it was also reported that resistance was observed in the treatment of trametinib, suggesting that different MEK1 mutations may have different response to trametinib, and further studies are necessary to distinguish that which MEK1 mutations are appropriate for the treatment with trametinib and which are not.
Collapse
Affiliation(s)
- Tong Lian
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Changying Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Haitao Wang
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Department of Oncology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
81
|
Satou A, Bennani NN, Feldman AL. Update on the classification of T-cell lymphomas, Hodgkin lymphomas, and histiocytic/dendritic cell neoplasms. Expert Rev Hematol 2019; 12:833-843. [PMID: 31365276 DOI: 10.1080/17474086.2019.1647777] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: The classification of lymphomas is based on the postulated normal counterparts of lymphoid neoplasms and currently constitutes over 100 definite or provisional entities. As this number of entities implies, lymphomas show marked pathological, genetic, and clinical heterogeneity. Recent molecular findings have significantly advanced our understanding of lymphomas. Areas covered: The World Health Organization (WHO) classification of lymphoid neoplasms was updated in 2017. The present review summarizes the new findings that have been gained in the areas of mature T-cell neoplasms, Hodgkin lymphomas, and histiocytic/dendritic cell neoplasms since the publication of the 2017 WHO classification. Expert opinion: Although formal revisions to the WHO classification are published only periodically, our understanding of the pathologic, genetic, and clinical features of lymphoid neoplasms is constantly evolving, particularly in the age of -omics technologies and targeted therapeutics. Even in the relatively short time since the publication of the 2017 WHO classification, many significant findings have been identified in the entities covered in this review.
Collapse
Affiliation(s)
- Akira Satou
- Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester , MN , USA.,Department of Surgical Pathology, Aichi Medical University Hospital , Nagakute , Aichi , Japan
| | - N Nora Bennani
- Division of Hematology, Mayo Clinic , Rochester , MN , USA
| | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
82
|
Héritier S, Emile JF, Hélias-Rodzewicz Z, Donadieu J. Progress towards molecular-based management of childhood Langerhans cell histiocytosis. Arch Pediatr 2019; 26:301-307. [PMID: 31281037 DOI: 10.1016/j.arcped.2019.05.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/13/2019] [Accepted: 05/30/2019] [Indexed: 12/13/2022]
Abstract
Langerhans cell histiocytosis (LCH) is characterized by inflammatory lesions containing abundant CD1a+ CD207+ histiocytes that lead to the destruction of affected tissues. This disease has a remarkable pleiotropic clinical presentation and most commonly affects young children. Although the current mortality rate is very low for childhood LCH patients (<2%), reactivation frequently occurs after a long period of disease control and the rates of permanent complications and sequelae remain high. Advances in genomic sequencing technologies in this past decade have highlighted somatic molecular alterations responsible for the disease in around 80% of childhood LCH cases. More than half of these cases harbored the BRAFV600E mutation, and most other mutations also concerned proteins involved in the MAPKinase pathway. In addition to improving what is known about the LCH pathology, this molecular knowledge provides opportunities to optimize patient management. The BRAFV600E mutation is associated with more severe presentations of the disease, a high reactivation rate, and a high permanent complication rate; this mutation therefore paves the way for future stratified management approaches. These therapies may be based on the patient's molecular status as well as other clinical characteristics of the disease that are independently associated with undesired events. Moreover, as observed in patients with solid tumors, the BRAFV600E allele can be detected in the circulating cell-free DNA of patients with severe BRAFV600E-mutated LCH. Quantification of the plasmatic BRAFV600E load for this group of patients can precisely monitor response to therapy. Finally, targeted therapies, such as BRAF inhibitors, are new therapeutic options especially designed for refractory multisystemic LCH involving risk organs. However, the long-term efficacy, long-term tolerance, optimal protocol scheme, and appropriate modalities of administration for these innovative therapies for children still need to be defined, a huge challenge.
Collapse
Affiliation(s)
- S Héritier
- Faculté de médecine, Sorbonne Université, 75013 Paris, France; Service d'hématologie oncologie pédiatrique, centre de référence des histiocytoses, hôpital Armand-Trousseau, Assistance publique-Hôpitaux de Paris, 6, avenue du Dr Netter, 75012 Paris, France; EA4340, UVSQ, université Paris-Saclay, 92100 Boulogne-Billancourt, France.
| | - J-F Emile
- EA4340, UVSQ, université Paris-Saclay, 92100 Boulogne-Billancourt, France; Département de pathologie, hôpital Ambroise-Paré, Assistance publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France
| | - Z Hélias-Rodzewicz
- EA4340, UVSQ, université Paris-Saclay, 92100 Boulogne-Billancourt, France; Département de pathologie, hôpital Ambroise-Paré, Assistance publique-Hôpitaux de Paris, 92104 Boulogne-Billancourt, France
| | - J Donadieu
- Service d'hématologie oncologie pédiatrique, centre de référence des histiocytoses, hôpital Armand-Trousseau, Assistance publique-Hôpitaux de Paris, 6, avenue du Dr Netter, 75012 Paris, France; EA4340, UVSQ, université Paris-Saclay, 92100 Boulogne-Billancourt, France
| |
Collapse
|
83
|
Papo M, Cohen-Aubart F, Trefond L, Bauvois A, Amoura Z, Emile JF, Haroche J. Systemic Histiocytosis (Langerhans Cell Histiocytosis, Erdheim-Chester Disease, Destombes-Rosai-Dorfman Disease): from Oncogenic Mutations to Inflammatory Disorders. Curr Oncol Rep 2019; 21:62. [PMID: 31115724 DOI: 10.1007/s11912-019-0810-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Provide an overview of recent progress in decoding the pathogenesis and treatment of systemic histiocytoses. RECENT FINDINGS Advances in molecular techniques over the last few years, enabling the identification of several MAPK mutations in lesion histiocytes, have revolutionized our understanding of histiocytosis that led to a revised classification and new treatments. Since the 2010 discovery of the BRAFV600E mutation in 57% of Langerhans cell histiocytosis (LCH) lesions, several other kinase mutations have been found, mostly in the MAPK pathway, and also in other key signaling pathways, in LCH, Erdheim-Chester Disease (ECD) and, less frequently, Destombes-Rosai-Dorfman disease (RDD). Those revolutionary breakthroughs enhanced our understanding of the pathogenesis of histiocytosis and led to trials with targeted therapies that demonstrated notable efficacy.
Collapse
Affiliation(s)
- Matthias Papo
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne 2, Centre National de Références des Histiocytoses, Hôpital Pitié-Salpêtrière, Sorbonne Université, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Fleur Cohen-Aubart
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne 2, Centre National de Références des Histiocytoses, Hôpital Pitié-Salpêtrière, Sorbonne Université, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Ludovic Trefond
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne 2, Centre National de Références des Histiocytoses, Hôpital Pitié-Salpêtrière, Sorbonne Université, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Adeline Bauvois
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne 2, Centre National de Références des Histiocytoses, Hôpital Pitié-Salpêtrière, Sorbonne Université, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Zahir Amoura
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne 2, Centre National de Références des Histiocytoses, Hôpital Pitié-Salpêtrière, Sorbonne Université, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Jean-François Emile
- EA4340-BECCOH, Versailles University, & Département de Pathologie, Hôpital Ambroise Paré, AP-HP, 9 Avenue Charles de Gaulle, 92100, Boulogne, France
| | - Julien Haroche
- Assistance Publique-Hôpitaux de Paris, Service de Médecine Interne 2, Centre National de Références des Histiocytoses, Hôpital Pitié-Salpêtrière, Sorbonne Université, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France.
| |
Collapse
|
84
|
Maggio A, Climent F, Turégano P, Sirvent JJ. [Multisystem histiocytosis of Langerhans cells associated with Lymphomatoid papulosis: An accidental finding? Case report and literature review]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2019; 52:130-135. [PMID: 30902378 DOI: 10.1016/j.patol.2018.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/24/2018] [Accepted: 04/29/2018] [Indexed: 10/28/2022]
Abstract
Langerhans cell histiocytosis (LCH) is a disease characterized by proliferation of CD1a+dendritic cells with local or diffuse organ compromise. The identification of recurrent gene mutations has confirmed the hypothesis of LCH as a true neoplasm. Lymphomatoid papulosis (LyP) belongs to the spectrum of CD30+primary cutaneous lymphomas. LCH has been described in association with other lymphoproliferative disorders. However, lesions constituted by Langerhans cells (LC) have been commonly considered reactive, related to cytokines produced by the lymphoma-microenvironment interaction. Some authors designate these lesions as "Langerhans cells-like lesions". We present the case of a 28-years-old woman with multisystem LCH and simultaneous PyL lesions with reactive LC hyperplasia.
Collapse
Affiliation(s)
- Antonio Maggio
- Servicio de Anatomía Patológica, Hospital Universitario Joan XXIII, Tarragona, España.
| | - Fina Climent
- Servicio de Anatomía Patológica, Hospital Universitario de Bellvitge-IBIDELLL, L'Hospitalet de Llobregat , España
| | - Pilar Turégano
- Servicio de Dermatología, Hospital Universitario Joan XXIII, Tarragona, España
| | - Juan Jose Sirvent
- Servicio de Anatomía Patológica, Hospital Universitario Joan XXIII, Tarragona, España; Universidad Rovira i Virgili, Tarragona, España; Institut d'Investigació Sanitària Pere Virgili, Tarragona, España
| |
Collapse
|
85
|
CyclinD1 Is Useful to Differentiate Langerhans Cell Histiocytosis From Reactive Langerhans Cells. Am J Dermatopathol 2019; 41:188-192. [DOI: 10.1097/dad.0000000000001250] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
86
|
Diamond EL, Durham BH, Ulaner GA, Drill E, Buthorn J, Ki M, Bitner L, Cho H, Young RJ, Francis JH, Rampal R, Lacouture M, Brody LA, Ozkaya N, Dogan A, Rosen N, Iasonos A, Abdel-Wahab O, Hyman DM. Efficacy of MEK inhibition in patients with histiocytic neoplasms. Nature 2019; 567:521-524. [PMID: 30867592 PMCID: PMC6438729 DOI: 10.1038/s41586-019-1012-y] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 12/16/2022]
Abstract
Histiocytic neoplasms are a heterogeneous group of clonal haematopoietic disorders that are marked by diverse mutations in the mitogen-activated protein kinase (MAPK) pathway1,2. For the 50% of patients with histiocytosis who have BRAFV600 mutations3-5, RAF inhibition is highly efficacious and has markedly altered the natural history of the disease6,7. However, no standard therapy exists for the remaining 50% of patients who lack BRAFV600 mutations. Although ERK dependence has been hypothesized to be a consistent feature across histiocytic neoplasms, this remains clinically unproven and many of the kinase mutations that are found in patients who lack BRAFV600 mutations have not previously been biologically characterized. Here we show ERK dependency in histiocytoses through a proof-of-concept clinical trial of cobimetinib, an oral inhibitor of MEK1 and MEK2, in patients with histiocytoses. Patients were enrolled regardless of their tumour genotype. In parallel, MAPK alterations that were identified in treated patients were characterized for their ability to activate ERK. In the 18 patients that we treated, the overall response rate was 89% (90% confidence interval of 73-100). Responses were durable, with no acquired resistance to date. At one year, 100% of responses were ongoing and 94% of patients remained progression-free. Cobimetinib treatment was efficacious regardless of genotype, and responses were observed in patients with ARAF, BRAF, RAF1, NRAS, KRAS, MEK1 (also known as MAP2K1) and MEK2 (also known as MAP2K2) mutations. Consistent with the observed responses, the characterization of the mutations that we identified in these patients confirmed that the MAPK-pathway mutations were activating. Collectively, these data demonstrate that histiocytic neoplasms are characterized by a notable dependence on MAPK signalling-and that they are consequently responsive to MEK inhibition. These results extend the benefits of molecularly targeted therapy to the entire spectrum of patients with histiocytosis.
Collapse
Affiliation(s)
- Eli L Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Benjamin H Durham
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gary A Ulaner
- Weill Cornell Medical College, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Esther Drill
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin Buthorn
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michelle Ki
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lillian Bitner
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hana Cho
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Young
- Weill Cornell Medical College, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jasmine H Francis
- Ophthalmic Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raajit Rampal
- Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mario Lacouture
- Weill Cornell Medical College, New York, NY, USA
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynn A Brody
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neval Ozkaya
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Ahmet Dogan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neal Rosen
- Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexia Iasonos
- Weill Cornell Medical College, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Weill Cornell Medical College, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - David M Hyman
- Weill Cornell Medical College, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
87
|
Durham BH. Molecular characterization of the histiocytoses: Neoplasia of dendritic cells and macrophages. Semin Cell Dev Biol 2019. [DOI: 10.1016/j.semcdb.2018.03.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
88
|
Langerhans Cell Histiocytoma: A Benign Histiocytic Neoplasm of Diverse Lines of Terminal Differentiation. Am J Dermatopathol 2019; 41:29-36. [DOI: 10.1097/dad.0000000000001255] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
89
|
Phillips JJ, Gong H, Chen K, Joseph NM, van Ziffle J, Bastian BC, Grenert JP, Kline CN, Mueller S, Banerjee A, Nicolaides T, Gupta N, Berger MS, Lee HS, Pekmezci M, Tihan T, Bollen AW, Perry A, Shieh JT, Solomon DA. The genetic landscape of anaplastic pleomorphic xanthoastrocytoma. Brain Pathol 2019; 29:85-96. [PMID: 30051528 PMCID: PMC7837273 DOI: 10.1111/bpa.12639] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/26/2018] [Indexed: 12/30/2022] Open
Abstract
Pleomorphic xanthoastrocytoma (PXA) is an astrocytic neoplasm that is typically well circumscribed and can have a relatively favorable prognosis. Tumor progression to anaplastic PXA (WHO grade III), however, is associated with a more aggressive biologic behavior and worse prognosis. The factors that drive anaplastic progression are largely unknown. We performed comprehensive genomic profiling on a set of 23 PXAs from 19 patients, including 15 with anaplastic PXA. Four patients had tumor tissue from multiple recurrences, including two with anaplastic progression. We find that PXAs are genetically defined by the combination of CDKN2A biallelic inactivation and RAF alterations that were present in all 19 cases, most commonly as CDKN2A homozygous deletion and BRAF p.V600E mutation but also occasionally BRAF or RAF1 fusions or other rearrangements. The third most commonly altered gene in anaplastic PXA was TERT, with 47% (7/15) harboring TERT alterations, either gene amplification (n = 2) or promoter hotspot mutation (n = 5). In tumor pairs analyzed before and after anaplastic progression, two had increased copy number alterations and one had TERT promoter mutation at recurrence. Less commonly altered genes included TP53, BCOR, BCORL1, ARID1A, ATRX, PTEN, and BCL6. All PXA in this cohort were IDH and histone H3 wildtype, and did not contain alterations in EGFR. Genetic profiling performed on six regions from the same tumor identified intratumoral genomic heterogeneity, likely reflecting clonal evolution during tumor progression. Overall, anaplastic PXA is characterized by the combination of CDKN2A biallelic inactivation and oncogenic RAF kinase signaling as well as a relatively small number of additional genetic alterations, with the most common being TERT amplification or promoter mutation. These data define a distinct molecular profile for PXA and suggest additional genetic alterations, including TERT, may be associated with anaplastic progression.
Collapse
Affiliation(s)
- Joanna J. Phillips
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
| | - Henry Gong
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
| | - Katharine Chen
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
| | - Nancy M. Joseph
- Clinical Cancer Genomics LaboratoryUniversity of California San FranciscoSan FranciscoCA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCA
| | - Jessica van Ziffle
- Clinical Cancer Genomics LaboratoryUniversity of California San FranciscoSan FranciscoCA
| | - Boris C. Bastian
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
- Clinical Cancer Genomics LaboratoryUniversity of California San FranciscoSan FranciscoCA
| | - James P. Grenert
- Clinical Cancer Genomics LaboratoryUniversity of California San FranciscoSan FranciscoCA
- Department of PathologyUniversity of California San FranciscoSan FranciscoCA
| | - Cassie N. Kline
- Division of Pediatric Hematology/OncologyDepartment of PediatricsUniversity of California San FranciscoSan FranciscoCA
| | - Sabine Mueller
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
- Division of Pediatric Hematology/OncologyDepartment of PediatricsUniversity of California San FranciscoSan FranciscoCA
| | - Anuradha Banerjee
- Division of Pediatric Hematology/OncologyDepartment of PediatricsUniversity of California San FranciscoSan FranciscoCA
| | - Theodore Nicolaides
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
- Division of Pediatric Hematology/OncologyDepartment of PediatricsUniversity of California San FranciscoSan FranciscoCA
| | - Nalin Gupta
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
| | - Mitchel S. Berger
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
| | - Han S. Lee
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
| | - Melike Pekmezci
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
| | - Tarik Tihan
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
| | - Andrew W. Bollen
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
| | - Arie Perry
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCA
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
| | - Joseph T.C. Shieh
- Department of PediatricsDivision of Medical GeneticsUniversity of California San FranciscoSan FranciscoCA
- Institute for Human GeneticsUniversity of CaliforniaSan FranciscoCA
| | - David A. Solomon
- Division of NeuropathologyDepartment of PathologyUniversity of California San FranciscoSan FranciscoCA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California San FranciscoSan FranciscoCA
- Clinical Cancer Genomics LaboratoryUniversity of California San FranciscoSan FranciscoCA
| |
Collapse
|
90
|
Cai J, Huang X, Yin M, Pan C, Song L, Zhan Z, Chen J, Gao Y, Tang J, Li Y, Shen S. A novel fusion gene PLEKHA6-NTRK3 in langerhans cell histiocytosis. Int J Cancer 2019; 144:117-124. [PMID: 30098202 PMCID: PMC6587734 DOI: 10.1002/ijc.31636] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/17/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022]
Abstract
Langerhans cell histiocytosis (LCH) is the most common histiocytosis with constitutive activation of the RAS-RAF-MEK-ERK (MAPKinase) cell signaling pathway. We analyzed 89 cases of BRAF and MAP2K1 mutations by Sanger sequencing, of which 18 cases showed that these two gene mutations are negative. Whole genome sequencing of suitable specimens in these negative cases revealed a translocation from the 3 intron of PLEKHA6 to the 13 intron of NTRK3 in one case. We identified that this translocation could cause a novel fusion mutation, PLEKHA6-NTRK3. Overexpression of the PLEKHA6-NTRK3 mutant in NIH 3T3 cells enhanced MAPKinase pathway activation, promote cell growth. Our result suggested that a new mutation need be included in LCH molecular screening panel to better define its prevalence in LCH.
Collapse
Affiliation(s)
- Jiaoyang Cai
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Xia Huang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Minzhi Yin
- Department of Pathology, Shanghai Children's Medical CenterSchool of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Ci Pan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Lili Song
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Zhiyan Zhan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Jing Chen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Yijin Gao
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Jingyan Tang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Yanxin Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| | - Shuhong Shen
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Department of Hematology & OncologyPediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong UniversityShanghai200127China
| |
Collapse
|
91
|
Kobayashi M, Tojo A. Langerhans cell histiocytosis in adults: Advances in pathophysiology and treatment. Cancer Sci 2018; 109:3707-3713. [PMID: 30281871 PMCID: PMC6272080 DOI: 10.1111/cas.13817] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/24/2018] [Accepted: 09/29/2018] [Indexed: 12/27/2022] Open
Abstract
Langerhans cell histiocytosis (LCH) is a rare systemic disorder characterized by the accumulation of CD1a+/Langerin+ LCH cells and wide-ranging organ involvement. Langerhans cell histiocytosis was formerly referred to as histiocytosis X, until it was renamed in 1987. Langerhans cell histiocytosis β was named for its morphological similarity to skin Langerhans cells. Studies have shown that LCH cells originate from myeloid dendritic cells rather than skin Langerhans cells. There has been significant debate regarding whether LCH should be defined as an immune disorder or a neoplasm. A breakthrough in understanding the pathogenesis of LCH occurred in 2010 when a gain-of-function mutation in BRAF (V600E) was identified in more than half of LCH patient samples. Studies have since reported that 100% of LCH cases show ERK phosphorylation, indicating that LCH is likely to be a clonally expanding myeloid neoplasm. Langerhans cell histiocytosis is now defined as an inflammatory myeloid neoplasm in the revised 2016 Histiocyte Society classification. Randomized trials and novel approaches have led to improved outcomes for pediatric patients, but no well-defined treatments for adult patients have been developed to date. Although LCH is not fatal in all cases, delayed diagnosis or treatment can result in serious impairment of organ function and decreased quality of life. This study summarizes recent advances in the pathophysiology and treatment of adult LCH, to raise awareness of this "orphan disease".
Collapse
Affiliation(s)
- Masayuki Kobayashi
- Division of Molecular TherapyAdvanced Clinical Research CenterInstitute of Medical ScienceThe University of TokyoTokyoJapan
| | - Arinobu Tojo
- Division of Molecular TherapyAdvanced Clinical Research CenterInstitute of Medical ScienceThe University of TokyoTokyoJapan
| |
Collapse
|
92
|
Park H, Nishino M, Hornick JL, Jacobsen ED. Imaging of Histiocytosis in the Era of Genomic Medicine. Radiographics 2018; 39:95-114. [PMID: 30500304 DOI: 10.1148/rg.2019180054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Histiocytosis describes a group of diseases that have long been considered enigmatic in the history of medicine. Recently, novel genomic analyses have identified somatic oncogenic driver mutations responsible for the pathogenesis of these entities. These discoveries have led to the recharacterization of histiocytoses as neoplastic diseases and have opened a new era of precision medicine approaches for treatment. The histiocytic disorders demonstrate a variety of imaging manifestations involving multiple organ systems, and radiologists play a major role in diagnosis and monitoring. An up-to-date knowledge of the novel genomic discoveries and their implications is essential for radiologists to understand the new approaches to treating histiocytic disorders and to contribute as key members of the multidisciplinary treatment team. This article provides a cutting-edge review of the novel concepts in histiocytosis, with a focus on recent genomic discoveries and precision medicine approaches to treating the disease, and describes imaging manifestations with correlative histologic and genomic findings, with an emphasis on adult-onset cases and uncommon subtypes. ©RSNA, 2018.
Collapse
Affiliation(s)
- Hyesun Park
- From the Departments of Radiology (H.P., M.N.), Pathology (J.L.H.), and Medical Oncology (E.D.J.), Brigham and Women's Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| | - Mizuki Nishino
- From the Departments of Radiology (H.P., M.N.), Pathology (J.L.H.), and Medical Oncology (E.D.J.), Brigham and Women's Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| | - Jason L Hornick
- From the Departments of Radiology (H.P., M.N.), Pathology (J.L.H.), and Medical Oncology (E.D.J.), Brigham and Women's Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| | - Eric D Jacobsen
- From the Departments of Radiology (H.P., M.N.), Pathology (J.L.H.), and Medical Oncology (E.D.J.), Brigham and Women's Hospital and Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215
| |
Collapse
|
93
|
Yuan J, Ng WH, Lam PYP, Wang Y, Xia H, Yap J, Guan SP, Lee ASG, Wang M, Baccarini M, Hu J. The dimer-dependent catalytic activity of RAF family kinases is revealed through characterizing their oncogenic mutants. Oncogene 2018; 37:5719-5734. [PMID: 29930381 PMCID: PMC6202329 DOI: 10.1038/s41388-018-0365-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022]
Abstract
Although extensively studied for three decades, the molecular mechanisms that regulate the RAF/MEK/ERK kinase cascade remain ambiguous. Recent studies identified the dimerization of RAF as a key event in the activation of this cascade. Here, we show that in-frame deletions in the β3-αC loop activate ARAF as well as BRAF and other oncogenic kinases by enforcing homodimerization. By characterizing these RAF mutants, we find that ARAF has less allosteric and catalytic activity than the other two RAF isoforms, which arises from its non-canonical APE motif. Further, these RAF mutants exhibit a strong oncogenic potential, and a differential inhibitor resistance that correlates with their dimer affinity. Using these unique mutants, we demonstrate that active RAFs, including the BRAF(V600E) mutant, phosphorylate MEK in a dimer-dependent manner. This study characterizes a special category of oncogenic kinase mutations, and elucidates the molecular basis that underlies the differential ability of RAF isoforms to stimulate MEK-ERK pathway. Further, this study reveals a unique catalytic feature of RAF family kinases that can be exploited to control their activities for cancer therapies.
Collapse
Affiliation(s)
- Jimin Yuan
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Wan Hwa Ng
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Paula Y P Lam
- Division of Cellular and Molecular Research, Singapore, Singapore
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore
| | - Yu Wang
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Hongping Xia
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Jiajun Yap
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Shou Ping Guan
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Ann S G Lee
- Division of Medical Sciences, National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore
- Office of Clinical & Academic Faculty Affairs, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Department of Physiology, National University of Singapore, 2 Medical Drive, 117597, Singapore, Singapore
| | - Mei Wang
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore
| | - Manuela Baccarini
- Max F. Perutz Laboratories, University of Vienna, Doktor-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Jiancheng Hu
- Division of Cellular and Molecular Research, Singapore, Singapore.
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore.
| |
Collapse
|
94
|
Mitchell JM, Berzins SP, Kannourakis G. A potentially important role for T cells and regulatory T cells in Langerhans cell histiocytosis. Clin Immunol 2018; 194:19-25. [DOI: 10.1016/j.clim.2018.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/27/2018] [Accepted: 06/15/2018] [Indexed: 12/11/2022]
|
95
|
Lorillon G, Jouenne F, Baroudjian B, de Margerie-Mellon C, Vercellino L, Meignin V, Lebbe C, Vassallo R, Mourah S, Tazi A. Response to Trametinib of a Pulmonary Langerhans Cell Histiocytosis Harboring a MAP2K1 Deletion. Am J Respir Crit Care Med 2018; 198:675-678. [DOI: 10.1164/rccm.201802-0275le] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
| | - Fanelie Jouenne
- Assistance Publique-Hôpitaux de ParisParis, France
- Université Paris DiderotParis, France
| | | | | | | | - Véronique Meignin
- Assistance Publique-Hôpitaux de ParisParis, France
- INSERM UMR_S1165Paris, France
| | - Celeste Lebbe
- Assistance Publique-Hôpitaux de ParisParis, France
- Université Paris DiderotParis, France
- INSERM U976Paris, France
| | | | - Samia Mourah
- Assistance Publique-Hôpitaux de ParisParis, France
- Université Paris DiderotParis, France
- INSERM U976Paris, France
| | - Abdellatif Tazi
- Assistance Publique-Hôpitaux de ParisParis, France
- Université Paris DiderotParis, France
- U1153 CRESSParis, France
| |
Collapse
|
96
|
Affiliation(s)
- Carl E Allen
- From the Texas Children's Cancer Center and the Department of Pediatrics, Baylor College of Medicine, Houston (C.E.A., K.L.M.); and the Department of Oncological Sciences, the Precision Immunology Institute, and the Tisch Cancer Institute, Icahn School of Medicine, New York (M.M.)
| | - Miriam Merad
- From the Texas Children's Cancer Center and the Department of Pediatrics, Baylor College of Medicine, Houston (C.E.A., K.L.M.); and the Department of Oncological Sciences, the Precision Immunology Institute, and the Tisch Cancer Institute, Icahn School of Medicine, New York (M.M.)
| | - Kenneth L McClain
- From the Texas Children's Cancer Center and the Department of Pediatrics, Baylor College of Medicine, Houston (C.E.A., K.L.M.); and the Department of Oncological Sciences, the Precision Immunology Institute, and the Tisch Cancer Institute, Icahn School of Medicine, New York (M.M.)
| |
Collapse
|
97
|
BRAF
mutation leading to central nervous system rosai-dorfman disease. Ann Neurol 2018; 84:147-152. [DOI: 10.1002/ana.25281] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 11/07/2022]
|
98
|
McClain KL, Picarsic J, Chakraborty R, Zinn D, Lin H, Abhyankar H, Scull B, Shih A, Phaik Har Lim K, Eckstein O, Lubega J, Peters TL, Olea W, Burke T, Ahmed N, John Hicks M, Tran B, Jones J, Dauser R, Jeng M, Baiocchi R, Schiff D, Goldman S, Heym KM, Wilson H, Carcamo B, Kumar A, Rodriguez-Galindo C, Whipple NS, Campbell P, Murdoch G, Kofler J, Heales S, Malone M, Woltjer R, Quinn JF, Orchard P, Kruer MC, Jaffe R, Manz MG, Lira SA, Williams Parsons D, Merad M, Man TK, Allen CE. CNS Langerhans cell histiocytosis: Common hematopoietic origin for LCH-associated neurodegeneration and mass lesions. Cancer 2018; 124:2607-2620. [PMID: 29624648 PMCID: PMC6289302 DOI: 10.1002/cncr.31348] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/29/2018] [Accepted: 02/14/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Central nervous system Langerhans cell histiocytosis (CNS-LCH) brain involvement may include mass lesions and/or a neurodegenerative disease (LCH-ND) of unknown etiology. The goal of this study was to define the mechanisms of pathogenesis that drive CNS-LCH. METHODS Cerebrospinal fluid (CSF) biomarkers including CSF proteins and extracellular BRAFV600E DNA were analyzed in CSF from patients with CNS-LCH lesions compared with patients with brain tumors and other neurodegenerative conditions. Additionally, the presence of BRAFV600E was tested in peripheral mononuclear blood cells (PBMCs) as well as brain biopsies from LCH-ND patients, and the response to BRAF-V600E inhibitor was evaluated in 4 patients with progressive disease. RESULTS Osteopontin was the only consistently elevated CSF protein in patients with CNS-LCH compared with patients with other brain pathologies. BRAFV600E DNA was detected in CSF of only 2/20 (10%) cases, both with LCH-ND and active lesions outside the CNS. However, BRAFV600E+ PBMCs were detected with significantly higher frequency at all stages of therapy in LCH patients who developed LCH-ND. Brain biopsies of patients with LCH-ND demonstrated diffuse perivascular infiltration by BRAFV600E+ cells with monocyte phenotype (CD14+ CD33+ CD163+ P2RY12- ) and associated osteopontin expression. Three of 4 patients with LCH-ND treated with BRAF-V600E inhibitor experienced significant clinical and radiologic improvement. CONCLUSION In LCH-ND patients, BRAFV600E+ cells in PBMCs and infiltrating myeloid/monocytic cells in the brain is consistent with LCH-ND as an active demyelinating process arising from a mutated hematopoietic precursor from which LCH lesion CD207+ cells are also derived. Therapy directed against myeloid precursors with activated MAPK signaling may be effective for LCH-ND. Cancer 2018;124:2607-20. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Kenneth L. McClain
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Jennifer Picarsic
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rikhia Chakraborty
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Daniel Zinn
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Howard Lin
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Harshal Abhyankar
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Brooks Scull
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Albert Shih
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Karen Phaik Har Lim
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas
| | - Olive Eckstein
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Joseph Lubega
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Tricia L. Peters
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Department of Pathology, Baylor College of Medicine, Houston, Texas
| | - Walter Olea
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Thomas Burke
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Nabil Ahmed
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - M. John Hicks
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Department of Pathology, Baylor College of Medicine, Houston, Texas
| | - Brandon Tran
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| | - Jeremy Jones
- Department of Radiology, Baylor College of Medicine, Houston, Texas
| | - Robert Dauser
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Michael Jeng
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California
| | - Robert Baiocchi
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Deborah Schiff
- Department of Pediatrics, University of California-San Diego, La Jolla, California
| | - Stanton Goldman
- Medical City Children’s Hospital, Dallas Texas and Texas Oncology, Pennsylvania
| | - Kenneth M. Heym
- Department of Pediatrics, Cook Children’s Medical Center, Fort Worth, Texas
| | - Harry Wilson
- Department of Pathology, Texas Tech University Health Sciences Center El Paso, El Paso, Texas
| | - Benjamin Carcamo
- Department of Pediatrics, Texas Tech University Health Sciences Center El Paso, El Paso, Texas
| | - Ashish Kumar
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | | | | | | | - Geoffrey Murdoch
- Department of Pathology, Division of Neuropathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Julia Kofler
- Department of Pathology, Division of Neuropathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Simon Heales
- Chemical Pathology, Great Ormond Street Hospital for Children, London, UK
| | - Marian Malone
- Laboratory Medicine, Great Ormond Street Hospital for Children, London, UK
| | - Randy Woltjer
- Layton Aging and Alzheimer’s Disease Center, Oregon Health and Science University, Portland, Oregon
| | - Joseph F. Quinn
- Layton Aging and Alzheimer’s Disease Center, Oregon Health and Science University, Portland, Oregon
| | - Paul Orchard
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Michael C. Kruer
- Barrow Neurological Institute, Phoenix Children’s Hospital; Child Health, Neurology & Genetics, University of Arizona College of Medicine, Phoenix, Arizona
| | - Ronald Jaffe
- Department of Pathology, Magee-Women’s Hospital of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Markus G. Manz
- Division of Hematology, University of Zurich, University Hospital Zurich, Zurich, Switzerland
| | - Sergio A. Lira
- Immunology Institute, Icahn School of Medicine, New York, New York
| | - D. Williams Parsons
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine, New York, New York
| | - Tsz-Kwong Man
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Carl E. Allen
- Texas Children’s Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
99
|
Langerhans cell histiocytosis in children. J Am Acad Dermatol 2018; 78:1047-1056. [DOI: 10.1016/j.jaad.2017.05.060] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/15/2017] [Accepted: 05/31/2017] [Indexed: 12/23/2022]
|
100
|
Peckham-Gregory EC, McClain KL, Allen CE, Scheurer ME, Lupo PJ. The role of parental and perinatal characteristics on Langerhans cell histiocytosis: characterizing increased risk among Hispanics. Ann Epidemiol 2018; 28:521-528. [PMID: 29724524 PMCID: PMC6054892 DOI: 10.1016/j.annepidem.2018.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/23/2018] [Accepted: 04/10/2018] [Indexed: 12/24/2022]
Abstract
PURPOSE Potential roles of inherited and environmental risk factors in pathogenesis of Langerhans cell histiocytosis (LCH), a myeloid neoplastic disorder, are undefined. We therefore evaluated the role of parental and perinatal factors on the risk of this childhood cancer. METHODS Information on LCH cases (n = 162) for the period 1995-2011 was obtained from the Texas Cancer Registry. Birth certificate controls were frequency-matched on year of birth at a ratio of 10:1 for the same period. Variables evaluated included parental age, race/ethnicity, size for gestational age, and birth order. Logistic regression was used to generate an adjusted odds ratio (aOR) and 95% confidence interval (CI) testing the association between each factor and LCH. RESULTS Few perinatal or parental factors were associated with LCH risk, with the exception of race/ethnicity. Mothers of Hispanic ethnicity were more likely to have children who developed LCH compared to non-Hispanic whites (aOR: 1.51; 95% CI: 1.02-2.25). This risk increased when both parents were Hispanic (aOR: 1.80; 95% CI: 1.13-2.87). Non-Hispanic black mothers were suggested as less likely to give birth to offspring who developed LCH compared to non-Hispanic whites (aOR: 0.50; 95% CI: 0.24-1.02). CONCLUSIONS LCH is characterized by somatic mutations in MAPK pathway genes in myeloid precursors. Increased risk for LCH in children of Hispanic parents suggests potential impact of inherited factors on LCH pathogenesis.
Collapse
Affiliation(s)
- Erin C Peckham-Gregory
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX; Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - Kenneth L McClain
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX; Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - Carl E Allen
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX; Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX; Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | - Michael E Scheurer
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX; Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX
| | - Philip J Lupo
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX; Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Houston, TX.
| |
Collapse
|