101
|
Maurer B, Kollmann S, Pickem J, Hoelbl-Kovacic A, Sexl V. STAT5A and STAT5B-Twins with Different Personalities in Hematopoiesis and Leukemia. Cancers (Basel) 2019; 11:E1726. [PMID: 31690038 PMCID: PMC6895831 DOI: 10.3390/cancers11111726] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/25/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factors STAT5A and STAT5B have essential roles in survival and proliferation of hematopoietic cells-which have been considered largely redundant. Mutations of upstream kinases, copy number gains, or activating mutations in STAT5A, or more frequently in STAT5B, cause altered hematopoiesis and cancer. Interfering with their activity by pharmacological intervention is an up-and-coming therapeutic avenue. Precision medicine requests detailed knowledge of STAT5A's and STAT5B's individual functions. Recent evidence highlights the privileged role for STAT5B over STAT5A in normal and malignant hematopoiesis. Here, we provide an overview on their individual functions within the hematopoietic system.
Collapse
Affiliation(s)
- Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Judith Pickem
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Andrea Hoelbl-Kovacic
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria
| |
Collapse
|
102
|
STAT3 Dysregulation in Mature T and NK Cell Lymphomas. Cancers (Basel) 2019; 11:cancers11111711. [PMID: 31684088 PMCID: PMC6896161 DOI: 10.3390/cancers11111711] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 10/25/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract: T cell lymphomas comprise a distinct class of non-Hodgkin's lymphomas, which include mature T and natural killer (NK) cell neoplasms. While each malignancy within this group is characterized by unique clinicopathologic features, dysregulation in the Janus tyrosine family of kinases/Signal transducer and activator of transcription (JAK/STAT) signaling pathway, specifically aberrant STAT3 activation, is a common feature among these lymphomas. The mechanisms driving dysregulation vary among T cell lymphoma subtypes and include activating mutations in upstream kinases or STAT3 itself, formation of oncogenic kinases which drive STAT3 activation, loss of negative regulators of STAT3, and the induction of a pro-tumorigenic inflammatory microenvironment. Constitutive STAT3 activation has been associated with the expression of targets able to increase pro-survival signals and provide malignant fitness. Patients with dysregulated STAT3 signaling tend to have inferior clinical outcomes, which underscores the importance of STAT3 signaling in malignant progression. Targeting of STAT3 has shown promising results in pre-clinical studies in T cell lymphoma lines, ex-vivo primary malignant patient cells, and in mouse models of disease. However, targeting this pleotropic pathway in patients has proven difficult. Here we review the recent contributions to our understanding of the role of STAT3 in T cell lymphomagenesis, mechanisms driving STAT3 activation in T cell lymphomas, and current efforts at targeting STAT3 signaling in T cell malignancies.
Collapse
|
103
|
Histone lysine methyltransferases in biology and disease. Nat Struct Mol Biol 2019; 26:880-889. [PMID: 31582846 DOI: 10.1038/s41594-019-0298-7] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/24/2022]
Abstract
The precise temporal and spatial coordination of histone lysine methylation dynamics across the epigenome regulates virtually all DNA-templated processes. A large number of histone lysine methyltransferase (KMT) enzymes catalyze the various lysine methylation events decorating the core histone proteins. Mutations, genetic translocations and altered gene expression involving these KMTs are frequently observed in cancer, developmental disorders and other pathologies. Therapeutic compounds targeting specific KMTs are currently being tested in the clinic, although overall drug discovery in the field is relatively underdeveloped. Here we review the biochemical and biological activities of histone KMTs and their connections to human diseases, focusing on cancer. We also discuss the scientific and clinical challenges and opportunities in studying KMTs.
Collapse
|
104
|
Wang F, Liu T, Wang L, Gu Z, Yang N, Luo L, Zhu C, Li M, Liu D, Gao C. Unmanipulated haploidentical peripheral blood stem cell transplantation for patients with Philadelphia-negative acute lymphoblastic leukaemia in first complete remission. Leuk Lymphoma 2019; 61:118-127. [PMID: 31519118 DOI: 10.1080/10428194.2019.1660965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Haploidentical peripheral blood stem cell transplantation (Haplo-PBSCT) is a promising treatment option for patients with Ph-negative acute lymphoblastic leukemia (ALL). In this study, we retrospectively analyzed data from Ph-negative ALL patients who underwent haplo-PBSCT during their first complete remission (CR1), and compared the long-term outcomes between the standard-risk and high-risk patients. The 3-year probability of relapse was 7.6% and 16.7% for the standard- and high-risk group (p = .274). The 3-year probability of disease-free survival (DFS) and overall survival (OS) for the standard-risk versus high-risk groups were 84.6% versus 50% (p = .0063) and 92.3% versus 61.1% (p = .046), respectively. Univariate analysis showed that a diagnosis of high risk with fusion/mutation genes were associated with worse outcomes, which was confirmed by multivariate analysis (p = .016). In summary, haplo-PBSCT may be a promising alternative for patients with Ph-negative ALL in CR1, although the fusion/mutation genes in high-risk patients may relatively impair the long-term efficacy compared with standard-risk patients.
Collapse
Affiliation(s)
- Feiyan Wang
- Medical School, Nankai University, Tianjin, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Tong Liu
- Inpatient Department, 66242 Army Hospital, Xilin Gol, China
| | - Li Wang
- Department of Hematology and Oncology, Chinese PLA 401 Hospital, Qingdao, China
| | - Zhenyang Gu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Nan Yang
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Lan Luo
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chengying Zhu
- Medical School, Nankai University, Tianjin, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Meng Li
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Daihong Liu
- Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Chunji Gao
- Medical School, Nankai University, Tianjin, China.,Department of Hematology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
105
|
Lim MS, Bailey NG, King RL, Piris M. Molecular Genetics in the Diagnosis and Biology of Lymphoid Neoplasms. Am J Clin Pathol 2019; 152:277-301. [PMID: 31278738 DOI: 10.1093/ajcp/aqz078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The 2017 Workshop of the Society for Hematopathology/European Association for Haematopathology reviewed the role of molecular genetics in the diagnosis and biology of lymphoid neoplasms. METHODS The Workshop Panel reviewed 82 cases. RESULTS Molecular genetic testing reveals alterations that expand the spectrum of diseases such as DUSP22 rearrangement in ALK-negative anaplastic large cell lymphoma, large B-cell lymphoma with IRF4 rearrangement, MYD88 mutations in B-cell lymphomas, Burkitt-like lymphoma with 11q aberrations, and diagnostic criteria for high-grade B-cell lymphomas. Therapeutic agents and natural tumor progression may be associated with transcriptional reprogramming that lead to transdifferentiation and lineage switch. CONCLUSIONS Application of emerging technical advances has revealed the complexity of genetic events in lymphomagenesis, progression, and acquired resistance to therapies. They also contribute to enhanced understanding of the biology of indolent vs aggressive behavior, clonal evolution, tumor progression, and transcriptional reprogramming associated with transdifferentiation events that may occur subsequent to therapy.
Collapse
Affiliation(s)
- Megan S Lim
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | | | | | - Miguel Piris
- Department of Pathology, Centro de Investigación Biomédica en Red de Oncología, Hospital Universitario Fundación Jimenez Diaz, Madrid, Spain
| |
Collapse
|
106
|
The histone methyltransferase Setd2 is indispensable for V(D)J recombination. Nat Commun 2019; 10:3353. [PMID: 31350389 PMCID: PMC6659703 DOI: 10.1038/s41467-019-11282-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 06/19/2019] [Indexed: 12/28/2022] Open
Abstract
The diverse repertoire of T cell receptors (TCR) and immunoglobulins is generated through the somatic rearrangement of respective V, D and J gene segments, termed V(D)J recombination, during early T or B cell development. However, epigenetic regulation of V(D)J recombination is still not fully understood. Here we show that the deficiency of Setd2, a histone methyltransferase that catalyzes lysine 36 trimethylation on histone 3 (H3K36me3) in mice, causes a severe developmental block of thymocytes at the CD4−CD8− DN3 stage. While H3K36me3 is normally enriched at the TCRβ locus, Setd2 deficiency reduces TCRβ H3K36me3 and suppresses TCRβ V(D)J rearrangement by impairing RAG1 binding to TCRβ loci and the DNA double-strand break repair. Similarly, Setd2 ablation also impairs immunoglobulin V(D)J rearrangement to induce B cell development block at the pro-B stage. Lastly, SETD2 is frequently mutated in patients with primary immunodeficiency. Our study thus demonstrates that Setd2 is required for optimal V(D)J recombination and normal lymphocyte development. The repertoire of adaptive immune receptor is generated by V(D)J recombination, somatic rearrangements of V, D and J gene segments, in the respective loci. Here the authors show that the deficiency of Setd2, a histone methyl transfer, impairs V(D)J recombination and induces severe developmental blocks in both T and B lineages.
Collapse
|
107
|
Zhang HH, Wang HS, Qian XW, Fan CQ, Li J, Miao H, Zhu XH, Yu Y, Meng JH, Cao P, Le J, Jiang JY, Jiang WJ, Wang P, Zhai XW. Genetic variants and clinical significance of pediatric acute lymphoblastic leukemia. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:296. [PMID: 31475166 DOI: 10.21037/atm.2019.04.80] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Acute lymphoblastic leukemia (ALL), the most common childhood malignancy, is characterized by molecular aberrations. Recently, genetic profiling has been fully investigated on ALL; however, the interaction between its genetic alterations and clinical features is still unclear. Therefore, we investigated the effects of genetic variants on ALL phenotypes and clinical outcomes. Methods Targeted exome sequencing technology was used to detect molecular profiling of 140 Chinese pediatric patients with ALL. Correlation of genetic features and clinical outcomes was analyzed. Results T-cell ALL (T-ALL) patients had higher initial white blood cell (WBC) count (34.8×109/L), higher incidence of mediastinal mass (26.9%), more relapse (23.1%), and enriched NOTCH1 (23.1%), FBXW7 (23.1%) and PHF6 (11.5%) mutations. Among the 18 recurrently mutated genes, SETD2 and TP53 mutations occurred more in female patients (P=0.041), NOTCH1 and SETD2 mutants were with higher initial WBC counts (≥50×109/L) (P=0.047 and P=0.041), JAK1 mutants were with higher minimal residual disease (MRD) level both on day 19 and day 46 (day 19 MRD ≥1%, P=0.039; day 46 MRD ≥0.01%, P=0.031) after induction chemotherapy. Multivariate analysis revealed that initial WBC counts (≥50×109/L), MLLr, and TP53 mutations were independent risk factors for 3-year relapse free survival (RFS) in ALL. Furthermore, TP53 mutations, age (<1 year or ≥10 years), and MLLr were independently associated with adverse outcome in B-cell ALL (B-ALL). Conclusions MLLr and TP53 mutations are powerful predictors for adverse outcome in pediatric B-ALL and ALL. Genetic profiling can contribute to the improvement of prognostication and management in ALL patients.
Collapse
Affiliation(s)
- Hong-Hong Zhang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Hong-Sheng Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiao-Wen Qian
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Cui-Qing Fan
- Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jun Li
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Hui Miao
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiao-Hua Zhu
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yi Yu
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jian-Hua Meng
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ping Cao
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jun Le
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Jun-Ye Jiang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wen-Jing Jiang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ping Wang
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiao-Wen Zhai
- Department of Hematology and Oncology, Children's Hospital of Fudan University, Shanghai 201102, China.,Department of Pediatrics, Children's Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
108
|
Leval L. Breast implant‐associated anaplastic large cell lymphoma and other rare T‐cell lymphomas. Hematol Oncol 2019; 37 Suppl 1:24-29. [DOI: 10.1002/hon.2582] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Laurence Leval
- Institute of PathologyLausanne University Hospital Lausanne Switzerland
| |
Collapse
|
109
|
Nagel S, Pommerenke C, MacLeod RAF, Meyer C, Kaufmann M, Fähnrich S, Drexler HG. Deregulated expression of NKL homeobox genes in T-cell lymphomas. Oncotarget 2019; 10:3227-3247. [PMID: 31143370 PMCID: PMC6524933 DOI: 10.18632/oncotarget.26929] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/29/2019] [Indexed: 11/25/2022] Open
Abstract
Recently, we have presented a scheme, termed "NKL-code", which describes physiological expression patterns of NKL homeobox genes in early hematopoiesis and in lymphopoiesis including main stages of T-, B- and NK-cell development. Aberrant activity of these genes underlies the generation of hematological malignancies notably T-cell leukemia. Here, we searched for deregulated NKL homeobox genes in main entities of T-cell lymphomas comprising angioimmunoblastic T-cell lymphoma (AITL), anaplastic large cell lymphoma (ALCL), adult T-cell leukemia/lymphoma (ATLL), hepatosplenic T-cell lymphoma (HSTL), NK/T-cell lymphoma (NKTL) and peripheral T-cell lymphoma (PTCL). Our data revealed altogether 19 aberrantly overexpressed genes in these types, demonstrating deregulated NKL homeobox genes involvement in T-cell lymphomas as well. For detailed analysis we focused on NKL homeobox gene MSX1 which is normally expressed in NK-cells. MSX1 was overexpressed in subsets of HSTL patients and HSTL-derived sister cell lines DERL-2 and DERL-7 which served as models to characterize mechanisms of deregulation. We performed karyotyping, genomic and expression profiling, and whole genome sequencing to reveal mutated and deregulated gene candidates, including the fusion gene CD53-PDGFRB. Subsequent knockdown experiments allowed the reconstruction of an aberrant network involved in MSX1 deregulation, including chromatin factors AUTS2 and mutated histone HIST1H3B(K27M). The gene encoding AUTS2 is located at chromosome 7q11 and may represent a basic target of the HSTL hallmark aberration i(7q). Taken together, our findings highlight an oncogenic role for deregulated NKL homeobox genes in T-cell lymphoma and identify MSX1 as a novel player in HSTL, implicated in aberrant NK- and T-cell differentiation.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Claudia Pommerenke
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Roderick A F MacLeod
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Corinna Meyer
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Maren Kaufmann
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Silke Fähnrich
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Hans G Drexler
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| |
Collapse
|
110
|
Wong K, Robles-Espinoza CD, Rodriguez D, Rudat SS, Puig S, Potrony M, Wong CC, Hewinson J, Aguilera P, Puig-Butille JA, Bressac-de Paillerets B, Zattara H, van der Weyden L, Fletcher CDM, Brenn T, Arends MJ, Quesada V, Newton-Bishop JA, Lopez-Otin C, Bishop DT, Harms PW, Johnson TM, Durham AB, Lombard DB, Adams DJ. Association of the POT1 Germline Missense Variant p.I78T With Familial Melanoma. JAMA Dermatol 2019; 155:604-609. [PMID: 30586141 PMCID: PMC6506889 DOI: 10.1001/jamadermatol.2018.3662] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 08/23/2018] [Indexed: 12/31/2022]
Abstract
Importance The protection of telomeres 1 protein (POT1) is a critical component of the shelterin complex, a multiple-protein machine that regulates telomere length and protects telomere ends. Germline variants in POT1 have been linked to familial melanoma, and somatic mutations are associated with a range of cancers including cutaneous T-cell lymphoma (CTCL). Objective To characterize pathogenic variation in POT1 in families with melanoma to inform clinical management. Design, Setting, and Participants In this case study and pedigree evaluation, analysis of the pedigree of 1 patient with melanoma revealed a novel germline POT1 variant (p.I78T, c.233T>C, chromosome 7, g.124870933A>G, GRCh38) that was subsequently found in 2 other pedigrees obtained from the GenoMEL Consortium. Main Outcomes and Measures (1) Identification of the POT1 p.I78T variant; (2) evaluation of the clinical features and characteristics of patients with this variant; (3) analysis of 3 pedigrees; (4) genomewide single-nucleotide polymorphism genotyping of germline DNA; and (5) a somatic genetic analysis of available nevi and 1 melanoma lesion. Results The POT1 p.I78T variant was found in 3 melanoma pedigrees, all of persons who self-reported as being of Jewish descent, and was shown to disrupt POT1-telomere binding. A UV mutation signature was associated with nevus and melanoma formation in POT1 variant carriers, and somatic mutations in driver genes such as BRAF, NRAS, and KIT were associated with lesion development in these patients. Conclusions and Relevance POT1 p.I78T is a newly identified, likely pathogenic, variant meriting screening for in families with melanoma after more common predisposition genes such as CDKN2A have been excluded. It could also be included as part of gene panel testing.
Collapse
Affiliation(s)
- Kim Wong
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, England
| | - Carla Daniela Robles-Espinoza
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, England
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Campus Juriquilla, Santiago de Querétaro, Qro, Mexico
| | - David Rodriguez
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Saskia S. Rudat
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, England
| | - Susana Puig
- Melanoma Unit, Department of Dermatology, Hospital Clínic de Barcelona, IDIBAPS, Barcelona University, Barcelona, Spain
- Centre of Biomedical Research on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Miriam Potrony
- Melanoma Unit, Department of Dermatology, Hospital Clínic de Barcelona, IDIBAPS, Barcelona University, Barcelona, Spain
- Centre of Biomedical Research on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Chi C. Wong
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, England
| | - James Hewinson
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, England
| | - Paula Aguilera
- Melanoma Unit, Department of Dermatology, Hospital Clínic de Barcelona, IDIBAPS, Barcelona University, Barcelona, Spain
- Centre of Biomedical Research on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Joan Anton Puig-Butille
- Centre of Biomedical Research on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
- Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Barcelona University, Barcelona, Spain
| | - Brigitte Bressac-de Paillerets
- Gustave Roussy, Université Paris-Saclay, Département de Biologie et Pathologie Médicales, Villejuif, France
- INSERM U1186, Université Paris-Saclay, Villejuif, France
| | - Hélène Zattara
- Département de Génétique, APHM, CHU Timone-Enfants, Marseille, France
| | | | | | - Thomas Brenn
- Pathology Department, Western General Hospital, Edinburgh, Scotland
| | - Mark J. Arends
- Pathology Department, Western General Hospital, Edinburgh, Scotland
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Julia A. Newton-Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, England
| | - Carlos Lopez-Otin
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Centro de Investigación en Red de Cáncer (CIBERONC), Madrid, Spain
| | - D. Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, England
| | - Paul W. Harms
- Department of Pathology, University of Michigan, Ann Arbor
| | | | | | | | - David J. Adams
- Experimental Cancer Genetics, The Wellcome Trust Sanger Institute, Hinxton, England
| |
Collapse
|
111
|
Butler RM, McKenzie RC, Jones CL, Flanagan CE, Woollard WJ, Demontis M, Ferreira S, Tosi I, John S, Whittaker SJ, Mitchell TJ. Contribution of STAT3 and RAD23B in Primary Sézary Cells to Histone Deacetylase Inhibitor FK228 Resistance. J Invest Dermatol 2019; 139:1975-1984.e2. [PMID: 30910759 DOI: 10.1016/j.jid.2019.03.1130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/23/2019] [Accepted: 03/06/2019] [Indexed: 01/12/2023]
Abstract
FK228 (romidepsin) and suberoylanilide hydroxamic acid (vorinostat) are histone deacetylase inhibitors (HDACi) approved by the US Food and Drug Administration for cutaneous T-cell lymphoma (CTCL), including the leukemic subtype Sézary syndrome. This study investigates RAD23B and STAT3 gene perturbations in a large cohort of primary Sézary cells and the effect of FK228 treatment on tyrosine phosphorylation of STAT3 (pYSTAT3) and RAD23B expression. We report RAD23B copy number variation in 10% (12/119, P ≤ 0.01) of SS patients, associated with reduced mRNA expression (P = 0.04). RAD23B knockdown in a CTCL cell line led to a reduction in FK228-induced apoptosis. Histone deacetylase inhibitor treatment significantly reduced pYSTAT3 in primary Sézary cells and was partially mediated by RAD23B. A distinct pattern of RAD23B-pYSTAT3 co-expression in primary Sézary cells was detected. Critically, Sézary cells harboring the common STAT3 Y640F variant were less sensitive to FK228-induced apoptosis and exogenous expression of STAT3 Y640F, and D661Y conferred partial resistance to STAT3 transcriptional inhibition by FK228 (P ≤ 0.0024). These findings suggest that RAD23B and STAT3 gene perturbations could reduce sensitivity to histone deacetylase inhibitors in SS patients.
Collapse
Affiliation(s)
- Rosie M Butler
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Robert C McKenzie
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Christine L Jones
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Charlotte E Flanagan
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Wesley J Woollard
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Maria Demontis
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Silvia Ferreira
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Isabella Tosi
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Susan John
- Department of Immunology, Infection and Inflammatory Disease, King's College London, Guy's Hospital, London, UK
| | - Sean J Whittaker
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - Tracey J Mitchell
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK.
| |
Collapse
|
112
|
Bergmann AK, Fataccioli V, Castellano G, Martin-Garcia N, Pelletier L, Ammerpohl O, Bergmann J, Bhat J, Pau ECDS, Martín-Subero JI, Moffitt AB, Valencia A, Oberg HH, Wesch D, Jayne S, Dyer MJS, Kabelitz D, Gaulard P, Siebert R. DNA methylation profiling of hepatosplenic T-cell lymphoma. Haematologica 2019; 104:e104-e107. [PMID: 30337361 PMCID: PMC6395348 DOI: 10.3324/haematol.2018.196196] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Anke K Bergmann
- Institute of Human Genetics, Christian-Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
- Department of Pediatrics, Christian-Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
- Institute of Human Genetics, Medical School Hannover (MHH), Germany
| | - Virginie Fataccioli
- Department of Pathology, AP-HP, Groupe hospitalier Henri Mondor; Université Paris-Est, Faculté de Médecine; Inserm U955, Créteil, France
| | - Giancarlo Castellano
- Departamento de Anatomía Patológica, Farmacología y Microbiología, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Spain
| | - Nadine Martin-Garcia
- Department of Pathology, AP-HP, Groupe hospitalier Henri Mondor; Université Paris-Est, Faculté de Médecine; Inserm U955, Créteil, France
| | - Laura Pelletier
- Department of Pathology, AP-HP, Groupe hospitalier Henri Mondor; Université Paris-Est, Faculté de Médecine; Inserm U955, Créteil, France
| | - Ole Ammerpohl
- Institute of Human Genetics, Christian-Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
- Institute of Human Genetics, University of Ulm & University Medical Center Ulm, Germany
| | - Juri Bergmann
- Anatomical Institute, Christian-Albrechts-University Kiel, Germany
| | - Jaydeep Bhat
- Institute for Immunology, Christian- Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | | | - José I Martín-Subero
- Departamento de Anatomía Patológica, Farmacología y Microbiología, Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Institució Catalana de Recerca i Estudis Avançats (ICREA), and Departament de Fonaments Clinics, Universitat de Barcelona, Spain
| | - Andrea B Moffitt
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC, USA
| | - Alfonso Valencia
- Barcelona Supercomputing Centre (BSC), Barcelona Spain
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain
| | - Hans-Heinrich Oberg
- Institute for Immunology, Christian- Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Daniela Wesch
- Institute for Immunology, Christian- Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Sandrine Jayne
- Ernest and Helen Scott Haematological Research Institute, University of Leicester, UK
| | - Martin J S Dyer
- Ernest and Helen Scott Haematological Research Institute, University of Leicester, UK
| | - Dieter Kabelitz
- Institute for Immunology, Christian- Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Philippe Gaulard
- Department of Pathology, AP-HP, Groupe hospitalier Henri Mondor; Université Paris-Est, Faculté de Médecine; Inserm U955, Créteil, France
| | - Reiner Siebert
- Institute of Human Genetics, Christian-Albrechts-University Kiel & University Hospital Schleswig-Holstein, Campus Kiel, Germany
- Institute of Human Genetics, University of Ulm & University Medical Center Ulm, Germany
| |
Collapse
|
113
|
Xavier AC, Suzuki R. Treatment and prognosis of mature (non-anaplastic) T- and NK-cell lymphomas in childhood, adolescents, and young adults. Br J Haematol 2019; 185:1086-1098. [PMID: 30706440 DOI: 10.1111/bjh.15772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Paediatric non-Hodgkin lymphomas (pNHL) are a diverse group of malignancies characterised by nodal and/or extranodal involvement. Less common pNHL forms include those derived from mature T- and natural killer (NK) cells. Much of our current understanding of paediatric mature (non-anaplastic) T/NK-cell lymphomas with respect to pathogenesis, diagnosis and treatment is extrapolated from adult literature. At the Sixth International Symposium on Childhood, Adolescent and Young Adult Non-Hodgkin Lymphoma, convened September 26-29, 2018 in Rotterdam, The Netherlands, some important aspects on diagnosis and outcomes of mature (non-anaplastic) T/NK-cell lymphoma in children and adolescents were discussed and will be reviewed in here.
Collapse
Affiliation(s)
- Ana C Xavier
- Division of Hematology/Oncology, Department of Pediatrics, Children's of Alabama/University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ritsuro Suzuki
- Department of Haematology/Oncology, Shimane University Hospital, Izumo, Japan
| |
Collapse
|
114
|
Brandt PH, Rahmat LT, Ali SS. A rare case of hepatosplenic gamma-delta T-cell lymphoma and secondary hemophagocytic lymphohistiocytosis. Clin Case Rep 2019; 7:164-169. [PMID: 30656034 PMCID: PMC6333078 DOI: 10.1002/ccr3.1924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/15/2018] [Accepted: 10/22/2018] [Indexed: 02/01/2023] Open
Abstract
Hepatosplenic gamma-delta T-cell lymphoma with concurrent hemophogocytic lymphohistiocytosis is a rare but well-recognized clinical scenario, associated with a grim prognosis. Clinicians must be aware of this aggressive type of lymphoma so that a prompt diagnosis can be made with timely initiation of systemic therapy and referral for bone marrow transplant.
Collapse
Affiliation(s)
- Philip H. Brandt
- Department of Hematology and Oncology, D’Amour Center for Cancer Care, Baystate Medical CenterUniversity of MassachusettsSpringfieldMassachusetts
| | - Leena T. Rahmat
- Department of OncologyJohns Hopkins University, Sidney Kimmel Comprehensive Cancer Center, Sibley Memorial HospitalWashingtonDistrict of Columbia
| | - Syed S. Ali
- Department of Hematology and Oncology, D’Amour Center for Cancer Care, Baystate Medical CenterUniversity of MassachusettsSpringfieldMassachusetts
| |
Collapse
|
115
|
Iqbal J, Amador C, McKeithan TW, Chan WC. Molecular and Genomic Landscape of Peripheral T-Cell Lymphoma. Cancer Treat Res 2019; 176:31-68. [PMID: 30596212 DOI: 10.1007/978-3-319-99716-2_2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Peripheral T-cell lymphoma (PTCL) is an uncommon group of lymphoma covering a diverse spectrum of entities. Little was known regarding the molecular and genomic landscapes of these diseases until recently but the knowledge is still quite spotty with many rarer types of PTCL remain largely unexplored. In this chapter, the recent findings from gene expression profiling (GEP) studies, including profiling data on microRNA, where available, will be presented with emphasis on the implication on molecular diagnosis, prognostication, and the identification of new entities (PTCL-GATA3 and PTCL-TBX21) in the PTCL-NOS group. Recent studies using next-generation sequencing have unraveled the mutational landscape in a number of PTCL entities leading to a marked improvement in the understanding of their pathogenesis and biology. While many mutations are shared among PTCL entities, the frequency varies and certain mutations are quite unique to a specific entity. For example, TET2 is often mutated but this is particularly frequent (70-80%) in angioimmunoblastic T-cell lymphoma (AITL) and IDH2 R172 mutations appear to be unique for AITL. In general, chromatin modifiers and molecular components in the CD28/T-cell receptor signaling pathways are frequently mutated. The major findings will be summarized in this chapter correlating with GEP data and clinical features where appropriate. The mutational landscape of cutaneous T-cell lymphoma, specifically on mycosis fungoides and Sezary syndrome, will also be discussed.
Collapse
Affiliation(s)
- Javeed Iqbal
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, US
| | - Catalina Amador
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, US
| | - Timothy W McKeithan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA
| | - Wing C Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
116
|
Abstract
Hepatosplenic T-cell lymphoma (HSTL) is a rare variant of extranodal peripheral T-cell lymphomas (PTCL), associated with aggressive disease course and a relentless track record for lethal outcomes. HSTL presents commonly in young men in their third or fourth decade. Of the known causes, immune dysregulation and immunosuppression are the key players in the pathogenesis of HSTL. Clinical manifestation includes hepatosplenomegaly, fevers, and weakness. Bone marrow involvement or organomegaly can cause cytopenias. Anthracycline-based regimens provide modest responses with most individuals dying within a year of diagnosis. Hematopoietic stem cell transplant (HSCT) can be offered to fit and eligible patients to prolong remissions. Disease relapse post chemotherapy has an aggressive phenotype, with limited salvage options available in the setting of declining performance status. Understanding the disease biology further to identify mechanistic-driven drug discovery could overcome the current limitations of existing therapeutic armamentarium.
Collapse
|
117
|
Siaghani PJ, Wong JT, Chan J, Weisenburger DD, Song JY. Epidemiology and Pathology of T- and NK-Cell Lymphomas. Cancer Treat Res 2019; 176:1-29. [PMID: 30596211 DOI: 10.1007/978-3-319-99716-2_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
PURPOSE This review will describe and update readers on the recent changes in the 2017 WHO classification regarding peripheral T-cell lymphomas. RECENT FINDINGS Signficant advances in molecular studies have resulted in revisions to the classification as well as introduction to provisional entities such as breast implant-associated ALCL and nodal PTCL with T-follicular helper phenotype. SUMMARY Major advances in molecular and gene expression profiling has expanded our knowledge of these rare and aggressive diseases.
Collapse
Affiliation(s)
- Parwiz J Siaghani
- Department of Pathology, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Jerry T Wong
- Department of Pathology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - John Chan
- Department of Pathology, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Dennis D Weisenburger
- Department of Pathology, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Joo Y Song
- Department of Pathology, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
118
|
Allen PB, Lechowicz MJ. Hematologic toxicity is rare in relapsed patients treated with belinostat: a systematic review of belinostat toxicity and safety in peripheral T-cell lymphomas. Cancer Manag Res 2018; 10:6731-6742. [PMID: 30584367 PMCID: PMC6289206 DOI: 10.2147/cmar.s149241] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) are an aggressive and diverse group of lymphomas with a T-cell origin. Most patients progress following initial treatment and require salvage therapy. The burden of symptoms is high due to its extra-nodal presentation, high rate of advanced disease, and associated cytopenias combined with its predilection for an elderly population. The disease is generally incurable at relapse in the absence of transplantation and treatment is aimed at prolonging life and reducing disease-related symptoms. Belinostat is a histone deacetylate inhibitor that was granted accelerated approval by the US Food and Drug Administration on July 3, 2014, for the treatment of relapsed PTCL. Here, a systemic review was conducted to assess the safety and efficacy of belinostat. A safety analysis involved 512 patients with relapsed malignancies, and an efficacy analysis focused on patients with relapsed PTCL and included a total of 144 patients. Common adverse events were noted including fatigue (35%), nausea (42.8%), and vomiting (28.5%), but comparatively low rates of grade 3/4 hematologic toxicity overall (6.4%). Efficacy analysis demonstrated an overall response rate of 25.7% and complete responses of 10.4% with the majority of discontinuations occurring for lack of efficacy. Ultimately, these results demonstrate that belinostat has comparable efficacy to other agents used in this setting and is well tolerated in regard to hematologic events, but there is limited data on patient-reported outcomes, reduction in disease-related symptoms, or quality of life.
Collapse
Affiliation(s)
- Pamela B Allen
- Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA,
| | - Mary Jo Lechowicz
- Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA,
| |
Collapse
|
119
|
Jacobsen ED, Weinstock DM. Challenges and implications of genomics for T-cell lymphomas. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:63-68. [PMID: 30504292 PMCID: PMC6246015 DOI: 10.1182/asheducation-2018.1.63] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Treatment outcomes for patients with peripheral T-cell lymphomas (PTCLs) and advanced-stage cutaneous T-cell lymphomas (CTCLs) remain poor. The past few years have witnessed an explosion in our understanding of the genetics of these diverse malignancies. Many subtypes harbor highly recurrent mutations, including single-nucleotide variants, insertions/deletions, and chromosomal rearrangements, that affect T-cell receptor signaling, costimulatory molecules, JAK/STAT and phosphatidylinositol 3-kinase pathways, transcription factors, and epigenetic modifiers. An important subset of these mutations is included within commercially available, multigene panels and, in rare circumstances, indicate therapeutic targets. However, current preclinical and clinical evidence suggests that only a minority of mutations identified in TCLs indicate biologic dependence. With a few exceptions that we highlight, mutations identified in TCLs should not be routinely used to select targeted therapies outside of a clinical trial. Participation in trials and publication of both positive and negative results remain the most important mechanisms for improving patient outcomes.
Collapse
MESH Headings
- Genomics/methods
- Humans
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Lymphoma, T-Cell, Peripheral/therapy
- Mutation
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Eric D. Jacobsen
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; and
| | - David M. Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; and
- Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
120
|
Matutes E. The 2017 WHO update on mature T- and natural killer (NK) cell neoplasms. Int J Lab Hematol 2018; 40 Suppl 1:97-103. [PMID: 29741263 DOI: 10.1111/ijlh.12817] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/01/2018] [Indexed: 01/07/2023]
Abstract
Over the last decade, there has been a significant body of information regarding the biology of the lymphoid neoplasms. This clearly supports the need for updating the 2008 WHO (World Health Organization) classification of haematopoietic and lymphoid tumours. The 2017 WHO classification is not a new edition but an update and revision of the 4th edition. New provisional entities but not new definitive entities are included, and novel molecular data in most of the entities and changes in the nomenclature in few of them have been incorporated. In the context of the mature T- and NK-cell neoplasms, the most relevant updates concern to: 1-dysregulation of the JAK/STAT pathway due to gene mutations which are common to various aggressive and indolent neoplasms; 2-incorporation of new molecular players that are relevant to the pathogenesis of these neoplasms and/or have prognostic implications; 3-inclusion of new provisional entities within the subgroups of anaplastic, primarily intestinal and cutaneous lymphomas such as breast implant-associated anaplastic large cell lymphoma, indolent T-cell lymphoproliferative disorder of the gastrointestinal tract and primary cutaneous acral CD8+ T-cell lymphoma; 4-identification of poor prognostic subtypes of peripheral T-cell lymphomas not otherwise specified (PTCL, NOS) characterized by overexpression of certain genes and of a subgroup PTCL, NOS with a T follicular phenotype that now is included together with angioimmunoblastic T-cell lymphoma under the umbrella of lymphomas with a T follicular helper phenotype; and 5-refinement on the designation and definition of already established entities. A review of the major changes will be outlined.
Collapse
Affiliation(s)
- E Matutes
- Haematopathology Unit, Hospital Clinic, Barcelona University, Barcelona, Spain
| |
Collapse
|
121
|
The Cooperative Relationship between STAT5 and Reactive Oxygen Species in Leukemia: Mechanism and Therapeutic Potential. Cancers (Basel) 2018; 10:cancers10100359. [PMID: 30262727 PMCID: PMC6210354 DOI: 10.3390/cancers10100359] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) are now recognized as important second messengers with roles in many aspects of signaling during leukemogenesis. They serve as critical cell signaling molecules that regulate the activity of various enzymes including tyrosine phosphatases. ROS can induce inactivation of tyrosine phosphatases, which counteract the effects of tyrosine kinases. ROS increase phosphorylation of many proteins including signal transducer and activator of transcription-5 (STAT5) via Janus kinases (JAKs). STAT5 is aberrantly activated through phosphorylation in many types of cancer and this constitutive activation is associated with cell survival, proliferation, and self-renewal. Such leukemic activation of STAT5 is rarely caused by mutation of the STAT5 gene itself but instead by overactive mutant receptors with tyrosine kinase activity as well as JAK, SRC family protein tyrosine kinases (SFKs), and Abelson murine leukemia viral oncogene homolog (ABL) kinases. Interestingly, STAT5 suppresses transcription of several genes encoding antioxidant enzymes while simultaneously enhancing transcription of NADPH oxidase. By doing so, STAT5 activation promotes an overall elevation of ROS level, which acts as a feed-forward loop, especially in high risk Fms-related tyrosine kinase 3 (FLT3) mutant leukemia. Therefore, efforts have been made recently to target ROS in cancer cells. Drugs that are able to either quench ROS production or inversely augment ROS-related signaling pathways both have potential as cancer therapies and may afford some selectivity by activating feedback inhibition of the ROS-STAT5 kinome. This review summarizes the cooperative relationship between ROS and STAT5 and explores the pros and cons of emerging ROS-targeting therapies that are selective for leukemia characterized by persistent STAT5 phosphorylation.
Collapse
|
122
|
Chen ES. Targeting epigenetics using synthetic lethality in precision medicine. Cell Mol Life Sci 2018; 75:3381-3392. [PMID: 30003270 PMCID: PMC11105276 DOI: 10.1007/s00018-018-2866-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/30/2018] [Accepted: 07/03/2018] [Indexed: 12/31/2022]
Abstract
Technological breakthroughs in genomics have had a significant impact on clinical therapy for human diseases, allowing us to use patient genetic differences to guide medical care. The "synthetic lethal approach" leverages on cancer-specific genetic rewiring to deliver a therapeutic regimen that preferentially targets malignant cells while sparing normal cells. The utility of this system is evident in several recent studies, particularly in poor prognosis cancers with loss-of-function mutations that become "treatable" when two otherwise discrete and unrelated genes are targeted simultaneously. This review focuses on the chemotherapeutic targeting of epigenetic alterations in cancer cells and consolidates a network that outlines the interplay between epigenetic and genetic regulators in DNA damage repair. This network consists of numerous synergistically acting relationships that are druggable, even in recalcitrant triple-negative breast cancer. This collective knowledge points to the dawn of a new era of personalized medicine.
Collapse
Affiliation(s)
- Ee Sin Chen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- National University Health System (NUHS), Singapore, 119228, Singapore.
- NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
123
|
|
124
|
McThenia SS, Rawwas J, Oliveira JL, Khan SP, Rodriguez V. Hepatosplenic γδ T-cell lymphoma of two adolescents: Case report and retrospective literature review in children, adolescents, and young adults. Pediatr Transplant 2018; 22:e13213. [PMID: 29921021 DOI: 10.1111/petr.13213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/02/2018] [Indexed: 01/25/2023]
Abstract
HSTCL is a highly aggressive malignancy with a poor prognosis. Case series and accounts have reported the use of different chemotherapy regimens with diverse patient outcomes. Most long-term survivors had undergone high-dose chemotherapy with autologous or allogeneic HCT. We describe two pediatric patients with HSTCL who were treated with chemotherapy followed by allogeneic HCT. Both patients are alive and in complete remission 2 and 8 years after therapy. Multiagent chemotherapy followed with allogeneic HCT seems to provide patients who have chemotherapy-sensitive disease a long-term disease-free survival.
Collapse
Affiliation(s)
- Sheila S McThenia
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jawhar Rawwas
- Minneapolis Children's Hospital, Minneapolis, MN, USA
| | | | - Shakila P Khan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA.,Pediatric Blood and Marrow Transplantation, Mayo Clinic, Rochester, MN, USA
| | - Vilmarie Rodriguez
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA.,Pediatric Blood and Marrow Transplantation, Mayo Clinic, Rochester, MN, USA.,Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
125
|
|
126
|
Molecular Insights Into Pathogenesis of Peripheral T Cell Lymphoma: a Review. Curr Hematol Malig Rep 2018; 13:318-328. [DOI: 10.1007/s11899-018-0460-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
127
|
Novel insights into the pathogenesis of T-cell lymphomas. Blood 2018; 131:2320-2330. [DOI: 10.1182/blood-2017-11-764357] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/09/2018] [Indexed: 12/11/2022] Open
Abstract
Abstract
T-cell lymphomas are a heterogeneous group of rare malignancies with overlapping clinical, immunologic, and histologic features. Recent advances in our understanding of T-cell differentiation based on gene expression profiling, next-generation sequencing, and transgenic mouse modeling studies have better elucidated the pathogenetic mechanisms underlying the diverse biology of T-cell lymphomas. These studies show that although genetic alterations in epigenetic modifiers are implicated in all subtypes of T-cell lymphomas, specific subtypes demonstrate enrichment for particular recurrent alterations targeting specific genes. In this regard, RHOA and TET2 alterations are prevalent in nodal T-cell lymphomas, particularly angioimmunoblastic T-cell lymphomas, peripheral T-cell lymphomas (PTCLs) not otherwise specified, and nodal PTCLs with T-follicular helper phenotype. JAK-STAT signaling pathways are mutationally activated in many extranodal T-cell lymphomas, such as natural killer/T-cell and hepatosplenic T-cell lymphomas. The functional significance of many of these genetic alterations is becoming better understood. Altogether these advances will continue to refine diagnostic criteria, improve prognostication, and identify novel therapeutic targets, resulting in improved outcomes for patient with T-cell lymphomas.
Collapse
|
128
|
Abstract
PURPOSE OF REVIEW We review the genetic foundations of different rare lymphomas to examine their shared origins. These data indicate the potential application of genomics to improve the diagnosis and treatment of these rare diseases. RECENT FINDINGS Next generation sequencing technologies have provided an important window into the genetic underpinnings of lymphomas. A growing body of evidence indicates that although some genetic alterations are specific to certain diseases, others are shared across different lymphomas. Many such genetic events have already demonstrated clinical utility, such as BRAF V600E that confers sensitivity to vemurafenib in patients with hairy cell leukemia. SUMMARY The rareness of many lymphoma subtypes makes the conduct of clinical trials and recruitment of significant numbers of patients impractical. However, a knowledge of the shared genetic origins of these rare lymphomas has the potential to inform 'basket' clinical trials in which multiple lymphoma subtypes are included. These trials would include patients based on the presence of alterations in targetable driver genes. Such approaches would be greatly strengthened by a systematic assessment of significant patient numbers from each subtype using next generation sequencing.
Collapse
|
129
|
Dual JAK1 and STAT3 mutations in a breast implant-associated anaplastic large cell lymphoma. Virchows Arch 2018; 473:505-511. [PMID: 29637270 DOI: 10.1007/s00428-018-2352-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/12/2018] [Accepted: 03/28/2018] [Indexed: 01/12/2023]
|
130
|
Greenplate A, Wang K, Tripathi RM, Palma N, Ali SM, Stephens PJ, Miller VA, Shyr Y, Guo Y, Reddy NM, Kozhaya L, Unutmaz D, Chen X, Irish JM, Davé UP. Genomic Profiling of T-Cell Neoplasms Reveals Frequent JAK1 and JAK3 Mutations With Clonal Evasion From Targeted Therapies. JCO Precis Oncol 2018; 2018. [PMID: 30079384 PMCID: PMC6072266 DOI: 10.1200/po.17.00019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose The promise of precision oncology is that identification of genomic alterations will direct the rational use of molecularly targeted therapy. This approach is particularly applicable to neoplasms that are resistant to standard cytotoxic chemotherapy, like T-cell leukemias and lymphomas. In this study, we tested the feasibility of targeted next-generation sequencing in profiles of diverse T-cell neoplasms and focused on the therapeutic utility of targeting activated JAK1 and JAK3 in an index case. Patients and Methods Using Foundation One and Foundation One Heme assays, we performed genomic profiling on 91 consecutive T-cell neoplasms for alterations in 405 genes. The samples were sequenced to high uniform coverage with an Illumina HiSeq and averaged a coverage depth of greater than 500× for DNA and more than 8M total pairs for RNA. An index case of T-cell prolymphocytic leukemia (T-PLL), which was analyzed by targeted next-generation sequencing, is presented. T-PLL cells were analyzed by RNA-seq, in vitro drug testing, mass cytometry, and phospho-flow. Results One third of the samples had genomic aberrations in the JAK-STAT pathway, most often composed of JAK1 and JAK3 gain-of-function mutations. We present an index case of a patient with T-PLL with a clonal JAK1 V658F mutation that responded to ruxolitinib therapy. After relapse developed, an expanded clone that harbored mutant JAK3 M511I and downregulation of the phosphatase, CD45, was identified. We demonstrate that the JAK missense mutations were activating, caused pathway hyperactivation, and conferred cytokine hypersensitivity. Conclusion These results underscore the utility of profiling occurrences of resistance to standard regimens and support JAK enzymes as rational therapeutic targets for T-cell leukemias and lymphomas.
Collapse
Affiliation(s)
| | - Kai Wang
- Foundation Medicine, Cambridge, MA. Origimed, Shanghai, China
| | | | | | | | | | | | - Yu Shyr
- Vanderbilt University Medical Center, Nashville, TN
| | - Yan Guo
- Vanderbilt University Medical Center, Nashville, TN
| | | | | | | | - Xueyan Chen
- University of Washington Medical Center, Seattle, WA
| | | | - Utpal P Davé
- R.L. Roudebush Veterans Affairs Medical Center and Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
131
|
Jordan J, Goldstein JS, Jaye DL, Gurcan M, Flowers CR, Cooper LAD. Informatics Approaches to Address New Challenges in the Classification of Lymphoid Malignancies. JCO Clin Cancer Inform 2018; 2. [PMID: 30637363 DOI: 10.1200/cci.17.00039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose Lymphoid malignancies are remarkably heterogeneous, with variations in outcomes and clinical, biologic, and histologic presentation complicating classification according to the World Health Organization guidelines. Incorrect classification of lymphoid neoplasms can result in suboptimal therapeutic strategies for individual patients and confound the interpretation of clinical trials involving personalized, class-based treatments. This review discusses the potential role of pathology informatics in improving the classification accuracy and objectivity for lymphoid malignancies. Design We identified peer-reviewed publications examining pathology informatics approaches for the classification of lymphoid malignancies, reviewed developments in the lymphoma classification systems, and summarized computational methods for pathologic assessment that can impact practice. Results Computer-assisted pathology image analysis algorithms in lymphoma most commonly have been applied to follicular lymphoma to address biologic heterogeneity and subjectivity in the process of classification. Conclusion Objective methods are available to assist pathologists in lymphoma classification and grading, and have been demonstrated to provide measurable benefits in specific contexts. Future validation and extension of these approaches will require datasets that link high resolution pathology images available for image analysis algorithms with clinical variables and follow up outcomes.
Collapse
Affiliation(s)
- Jacob Jordan
- Emory University School of Medicine, Atlanta, GA; Metin Gurcan, Ohio State University, Columbus, OH
| | - Jordan S Goldstein
- Emory University School of Medicine, Atlanta, GA; Metin Gurcan, Ohio State University, Columbus, OH
| | - David L Jaye
- Emory University School of Medicine, Atlanta, GA; Metin Gurcan, Ohio State University, Columbus, OH
| | - Metin Gurcan
- Emory University School of Medicine, Atlanta, GA; Metin Gurcan, Ohio State University, Columbus, OH
| | - Christopher R Flowers
- Emory University School of Medicine, Atlanta, GA; Metin Gurcan, Ohio State University, Columbus, OH
| | - Lee A D Cooper
- Emory University School of Medicine, Atlanta, GA; Metin Gurcan, Ohio State University, Columbus, OH
| |
Collapse
|
132
|
Nairismägi ML, Gerritsen ME, Li ZM, Wijaya GC, Chia BKH, Laurensia Y, Lim JQ, Yeoh KW, Yao XS, Pang WL, Bisconte A, Hill RJ, Bradshaw JM, Huang D, Song TLL, Ng CCY, Rajasegaran V, Tang T, Tang QQ, Xia XJ, Kang TB, Teh BT, Lim ST, Ong CK, Tan J. Oncogenic activation of JAK3-STAT signaling confers clinical sensitivity to PRN371, a novel selective and potent JAK3 inhibitor, in natural killer/T-cell lymphoma. Leukemia 2018; 32:1147-1156. [PMID: 29434279 PMCID: PMC5940653 DOI: 10.1038/s41375-017-0004-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 11/17/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023]
Abstract
Aberrant activation of the JAK3-STAT signaling pathway is a characteristic feature of many hematological malignancies. In particular, hyperactivity of this cascade has been observed in natural killer/T-cell lymphoma (NKTL) cases. Although the first-in-class JAK3 inhibitor tofacitinib blocks JAK3 activity in NKTL both in vitro and in vivo, its clinical utilization in cancer therapy has been limited by the pan-JAK inhibition activity. To improve the therapeutic efficacy of JAK3 inhibition in NKTL, we have developed a highly selective and durable JAK3 inhibitor PRN371 that potently inhibits JAK3 activity over the other JAK family members JAK1, JAK2, and TYK2. PRN371 effectively suppresses NKTL cell proliferation and induces apoptosis through abrogation of the JAK3-STAT signaling. Moreover, the activity of PRN371 has a more durable inhibition on JAK3 compared to tofacitinib in vitro, leading to significant tumor growth inhibition in a NKTL xenograft model harboring JAK3 activating mutation. These findings provide a novel therapeutic approach for the treatment of NKTL.
Collapse
Affiliation(s)
- M -L Nairismägi
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Z M Li
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - G C Wijaya
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - B K H Chia
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Y Laurensia
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - J Q Lim
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - K W Yeoh
- Department of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - X S Yao
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - W L Pang
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - A Bisconte
- Principia Biopharma, South San Francisco, CA, USA
| | - R J Hill
- Principia Biopharma, South San Francisco, CA, USA
| | - J M Bradshaw
- Principia Biopharma, South San Francisco, CA, USA
| | - D Huang
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - T L L Song
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - C C Y Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - V Rajasegaran
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - T Tang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Q Q Tang
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - X J Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - T B Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - B T Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - S T Lim
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Office of Education, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - C K Ong
- Lymphoma Genomic Translational Research Laboratory, Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore. .,Genome Institute of Singapore, A*STAR, Singapore, Singapore.
| | - J Tan
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore, Singapore. .,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
133
|
Yabe M, Miranda RN, Medeiros LJ. Hepatosplenic T-cell Lymphoma: a review of clinicopathologic features, pathogenesis, and prognostic factors. Hum Pathol 2018; 74:5-16. [PMID: 29337025 DOI: 10.1016/j.humpath.2018.01.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/28/2017] [Accepted: 01/02/2018] [Indexed: 12/31/2022]
Abstract
Hepatosplenic T-cell lymphoma (HSTCL) is a rare and clinically aggressive type of T-cell lymphoma that arises most often in adolescents and young adults. Patients with HSTCL commonly present with B-symptoms and cytopenias, which may suggest a diagnosis of acute leukemia initially. Patients present with extranodal disease involving the spleen, liver and bone marrow; lymphadenopathy is usually absent. The lymphoma cells can show a spectrum of cell sizes and are of T-cell lineage, often negative for CD4 and CD8 and positive for T-cell receptor γδ or, less often, αβ. Recent studies have identified gene mutations in oncogenic pathways that are likely involved in pathogenesis and may be targets for therapy. Mutations in STAT3 or STAT5B lead to activation of the JAK/STAT pathway, and mutations involving SETD2, IN080 and ARID1 are involved in chromatin modification. Currently, there is no consensus standard of care for HSTCL patients, although several studies support a role for allogeneic hematopoietic stem cell transplant. Although patients with HSTCL are best treated in the context of clinical trials, the rarity of these neoplasms likely necessitates a multi-institutional approach. In this review, we focus on the clinicopathologic and genetic characteristics of HSTCL. We also discuss the differential diagnosis and therapeutic approaches.
Collapse
Affiliation(s)
- Mariko Yabe
- Hematopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Roberto N Miranda
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
134
|
Abstract
<b/> In this issue of Cancer Discovery, McKinney and colleagues describe the genetics of hepatosplenic T-cell lymphoma, a rare subtype of T-cell lymphoma with unique clinical characteristics. The findings, specifically frequent mutations of STAT5B, PIK3CD, and the histone methyltransferase SETD2, may help guide translational efforts to target this deadly disease. Cancer Discov; 7(4); 352-3. ©2017 AACR.See related article by McKinney et al., p. 369.
Collapse
Affiliation(s)
- Noriaki Yoshida
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David M Weinstock
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Broad Institute of MIT and Harvard University, Cambridge, Massachusetts
| |
Collapse
|
135
|
Elenitoba-Johnson KSJ, Lim MS. New Insights into Lymphoma Pathogenesis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 13:193-217. [PMID: 29140757 DOI: 10.1146/annurev-pathol-020117-043803] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lymphomas represent clonal proliferations of lymphocytes that are broadly classified based upon their maturity (peripheral or mature versus precursor) and lineage (B cell, T cell, and natural killer cell). Insights into the pathogenetic mechanisms involved in lymphoma impact the classification of lymphoma and have significant implications for the diagnosis and clinical management of patients. Serial scientific and technologic advances over the last 30 years in immunology, cytogenetics, molecular biology, gene expression profiling, mass spectrometry-based proteomics, and, more recently, next-generation sequencing have contributed to greatly enhance our understanding of the pathogenetic mechanisms in lymphoma. Novel and emerging concepts that challenge our previously accepted paradigms about lymphoma biology and how these impact diagnosis, molecular testing, disease monitoring, drug development, and personalized and precision medicine for lymphoma are discussed.
Collapse
Affiliation(s)
- Kojo S J Elenitoba-Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; , .,Center for Personalized Diagnostics and Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; , .,Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
136
|
SETD2 alterations impair DNA damage recognition and lead to resistance to chemotherapy in leukemia. Blood 2017; 130:2631-2641. [PMID: 29018079 DOI: 10.1182/blood-2017-03-775569] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 09/18/2017] [Indexed: 02/06/2023] Open
Abstract
Mutations in SETD2, encoding the histone 3 lysine 36 trimethyltransferase, are enriched in relapsed acute lymphoblastic leukemia and MLL-rearranged acute leukemia. We investigated the impact of SETD2 mutations on chemotherapy sensitivity in isogenic leukemia cell lines and in murine leukemia generated from a conditional knockout of Setd2. SETD2 mutations led to resistance to DNA-damaging agents, cytarabine, 6-thioguanine, doxorubicin, and etoposide, but not to a non-DNA damaging agent, l-asparaginase. H3K36me3 localizes components of the DNA damage response (DDR) pathway and SETD2 mutation impaired DDR, blunting apoptosis induced by cytotoxic chemotherapy. Consistent with local recruitment of DDR, genomic regions with higher H3K36me3 had a lower mutation rate, which was increased with SETD2 mutation. Heterozygous conditional inactivation of Setd2 in a murine model decreased the latency of MLL-AF9-induced leukemia and caused resistance to cytarabine treatment in vivo, whereas homozygous loss delayed leukemia formation. Treatment with JIB-04, an inhibitor of the H3K9/36me3 demethylase KDM4A, restored H3K36me3 levels and sensitivity to cytarabine. These findings establish SETD2 alteration as a mechanism of resistance to DNA-damaging chemotherapy, consistent with a local loss of DDR, and identify a potential therapeutic strategy to target SETD2-mutant leukemias.
Collapse
|
137
|
New developments in the pathology of malignant lymphoma: a review of the literature published from January to April 2017. J Hematop 2017; 10:25-33. [PMID: 28819461 PMCID: PMC5537309 DOI: 10.1007/s12308-017-0295-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
138
|
Vallois D, Roberti A, Bisig B, Gaulard P, de Leval L. Du neuf dans l’oncogenèse moléculaire des lymphomes : rôle du gèneSETD2dans les lymphomes T de l’intestin. Med Sci (Paris) 2017; 33:469-473. [DOI: 10.1051/medsci/20173305004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
139
|
Diagnosis and classification of hematologic malignancies on the basis of genetics. Blood 2017; 130:410-423. [PMID: 28600336 DOI: 10.1182/blood-2017-02-734541] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Genomic analysis has greatly influenced the diagnosis and clinical management of patients affected by diverse forms of hematologic malignancies. Here, we review how genetic alterations define subclasses of patients with acute leukemias, myelodysplastic syndromes (MDS), myeloproliferative neoplasms (MPNs), non-Hodgkin lymphomas, and classical Hodgkin lymphoma. These include new subtypes of acute myeloid leukemia defined by mutations in RUNX1 or BCR-ABL1 translocations as well as a constellation of somatic structural DNA alterations in acute lymphoblastic leukemia. Among patients with MDS, detection of mutations in SF3B1 define a subgroup of patients with the ring sideroblast form of MDS and a favorable prognosis. For patients with MPNs, detection of the BCR-ABL1 fusion delineates chronic myeloid leukemia from classic BCR-ABL1- MPNs, which are largely defined by mutations in JAK2, CALR, or MPL In the B-cell lymphomas, detection of characteristic rearrangements involving MYC in Burkitt lymphoma, BCL2 in follicular lymphoma, and MYC/BCL2/BCL6 in high-grade B-cell lymphomas are essential for diagnosis. In T-cell lymphomas, anaplastic large-cell lymphoma is defined by mutually exclusive rearrangements of ALK, DUSP22/IRF4, and TP63 Genetic alterations affecting TP53 and the mutational status of the immunoglobulin heavy-chain variable region are important in clinical management of chronic lymphocytic leukemia. Additionally, detection of BRAFV600E mutations is helpful in the diagnosis of classical hairy cell leukemia and a number of histiocytic neoplasms. Numerous additional examples provided here demonstrate how clinical evaluation of genomic alterations have refined classification of myeloid neoplasms and major forms of lymphomas arising from B, T, or natural killer cells.
Collapse
|
140
|
Moffitt AB, Ondrejka SL, McKinney M, Rempel RE, Goodlad JR, Teh CH, Leppa S, Mannisto S, Kovanen PE, Tse E, Au-Yeung RKH, Kwong YL, Srivastava G, Iqbal J, Yu J, Naresh K, Villa D, Gascoyne RD, Said J, Czader MB, Chadburn A, Richards KL, Rajagopalan D, Davis NS, Smith EC, Palus BC, Tzeng TJ, Healy JA, Lugar PL, Datta J, Love C, Levy S, Dunson DB, Zhuang Y, Hsi ED, Dave SS. Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2. J Exp Med 2017; 214:1371-1386. [PMID: 28424246 PMCID: PMC5413324 DOI: 10.1084/jem.20160894] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 02/06/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022] Open
Abstract
Enteropathy-associated T cell lymphoma (EATL) is the most common oncologic complication of celiac disease. Moffitt and colleagues identify novel EATL-defining mutations in SETD2, as well as clinically relevant mutations in the JAK-STAT pathway. Enteropathy-associated T cell lymphoma (EATL) is a lethal, and the most common, neoplastic complication of celiac disease. Here, we defined the genetic landscape of EATL through whole-exome sequencing of 69 EATL tumors. SETD2 was the most frequently silenced gene in EATL (32% of cases). The JAK-STAT pathway was the most frequently mutated pathway, with frequent mutations in STAT5B as well as JAK1, JAK3, STAT3, and SOCS1. We also identified mutations in KRAS, TP53, and TERT. Type I EATL and type II EATL (monomorphic epitheliotropic intestinal T cell lymphoma) had highly overlapping genetic alterations indicating shared mechanisms underlying their pathogenesis. We modeled the effects of SETD2 loss in vivo by developing a T cell–specific knockout mouse. These mice manifested an expansion of γδ T cells, indicating novel roles for SETD2 in T cell development and lymphomagenesis. Our data render the most comprehensive genetic portrait yet of this uncommon but lethal disease and may inform future classification schemes.
Collapse
Affiliation(s)
- Andrea B Moffitt
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC 27708.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Sarah L Ondrejka
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Matthew McKinney
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Rachel E Rempel
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - John R Goodlad
- Haematological Malignancy Diagnostic Service, St. James's University Hospital, Leeds LS9 7TF, England, UK
| | - Chun Huat Teh
- Haematology Department, Western General Hospital, Edinburgh EH14 1TY, Scotland, UK
| | - Sirpa Leppa
- Department of Oncology and Research Program Unit, Faculty of Medicine, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Susanna Mannisto
- Department of Oncology and Research Program Unit, Faculty of Medicine, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Panu E Kovanen
- HUSLAB and Medicum, Helsinki University Hospital Cancer Center and University of Helsinki, 00014 Helsinki, Finland
| | - Eric Tse
- University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | | | - Yok-Lam Kwong
- University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | | | - Javeed Iqbal
- University of Nebraska Medical Center, Omaha, NE 68198
| | - Jiayu Yu
- University of Nebraska Medical Center, Omaha, NE 68198
| | | | - Diego Villa
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V6R 1ZE, Canada
| | - Randy D Gascoyne
- British Columbia Cancer Agency, University of British Columbia, Vancouver, BC V6R 1ZE, Canada
| | - Jonathan Said
- University of California, Los Angeles, Los Angeles, CA 90095
| | | | - Amy Chadburn
- Presbyterian Hospital, Pathology and Cell Biology, Cornell University, New York, NY 10065
| | | | | | - Nicholas S Davis
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Eileen C Smith
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Brooke C Palus
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Tiffany J Tzeng
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Jane A Healy
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Patricia L Lugar
- Department of Medicine, Duke University School of Medicine, Durham, NC 27710
| | - Jyotishka Datta
- Department of Statistical Science, Duke University, Durham, NC 27708
| | - Cassandra Love
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| | - Shawn Levy
- Hudson Alpha Institute for Biotechnology, Huntsville, AL 35806
| | - David B Dunson
- Department of Statistical Science, Duke University, Durham, NC 27708
| | - Yuan Zhuang
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710
| | - Eric D Hsi
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sandeep S Dave
- Duke Center for Genomics and Computational Biology, Duke University, Durham, NC 27708.,Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710
| |
Collapse
|
141
|
Marks LJ, Oberg JA, Pendrick D, Sireci AN, Glasser C, Coval C, Zylber RJ, Chung WK, Pang J, Turk AT, Hsiao SJ, Mansukhani MM, Glade Bender JL, Kung AL, Sulis ML. Precision Medicine in Children and Young Adults with Hematologic Malignancies and Blood Disorders: The Columbia University Experience. Front Pediatr 2017; 5:265. [PMID: 29312904 PMCID: PMC5732960 DOI: 10.3389/fped.2017.00265] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 11/28/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The advent of comprehensive genomic profiling has markedly advanced the understanding of the biology of pediatric hematological malignancies, however, its application to clinical care is still unclear. We present our experience integrating genomic data into the clinical management of children with high-risk hematologic malignancies and blood disorders and describe the broad impact that genomic profiling has in multiple aspects of patient care. METHODS The Precision in Pediatric Sequencing Program at Columbia University Medical Center instituted prospective clinical next-generation sequencing (NGS) for high-risk malignancies and blood disorders. Testing included cancer whole exome sequencing (WES) of matched tumor-normal samples or targeted sequencing of 467 cancer-associated genes, when sample adequacy was a concern, and tumor transcriptome (RNA-seq). A multidisciplinary molecular tumor board conducted interpretation of results and final tiered reports were transmitted to the electronic medical record according to patient preferences. RESULTS Sixty-nine samples from 56 patients with high-risk hematologic malignancies and blood disorders were sequenced. Patients carried diagnoses of myeloid malignancy (n = 25), lymphoid malignancy (n = 25), or histiocytic disorder (n = 6). Six patients had only constitutional WES, performed for a suspicion of an inherited predisposition for their disease. For the remaining 50 patients, tumor was sequenced with matched normal tissue when available. The mean number of somatic variants per sample was low across the different disease categories (2.85 variants/sample). Interestingly, a gene fusion was identified by RNA-seq in 58% of samples who had adequate RNA available for testing. Molecular profiling of tumor tissue led to clinically impactful findings in 90% of patients. Forty patients (80%) had at least one targetable gene variant or fusion identified in their tumor tissue; however, only seven received targeted therapy. Importantly, NGS findings contributed to the refinement of diagnosis and prognosis for 34% of patients. Known or likely pathogenic germline alterations were discovered in 24% of patients involving cancer predisposition genes in 12% of cases. CONCLUSION Incorporating whole exome and transcriptome profiling of tumor and normal tissue into clinical practice is feasible, and the value that comprehensive testing provides extends beyond the ability to target-specific mutations.
Collapse
Affiliation(s)
- Lianna J Marks
- Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jennifer A Oberg
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Columbia University Medical Center, New York, NY, United States
| | - Danielle Pendrick
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Columbia University Medical Center, New York, NY, United States
| | - Anthony N Sireci
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Chana Glasser
- Department of Pediatric Hematology/Oncology, NYU Winthrop University Medical Center, Mineola, NY, United States
| | - Carrie Coval
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States
| | - Rebecca J Zylber
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Columbia University Medical Center, New York, NY, United States
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, NY, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| | - Jiuhong Pang
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Andrew T Turk
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Susan J Hsiao
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Mahesh M Mansukhani
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| | - Julia L Glade Bender
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Columbia University Medical Center, New York, NY, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| | - Andrew L Kung
- Department of Pediatric Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Maria Luisa Sulis
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Columbia University Medical Center, New York, NY, United States.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|