1
|
Lees J, Hay J, Moles MW, Michie AM. The discrete roles of individual FOXO transcription factor family members in B-cell malignancies. Front Immunol 2023; 14:1179101. [PMID: 37275916 PMCID: PMC10233034 DOI: 10.3389/fimmu.2023.1179101] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Forkhead box (FOX) class O (FOXO) proteins are a dynamic family of transcription factors composed of four family members: FOXO1, FOXO3, FOXO4 and FOXO6. As context-dependent transcriptional activators and repressors, the FOXO family regulates diverse cellular processes including cell cycle arrest, apoptosis, metabolism, longevity and cell fate determination. A central pathway responsible for negative regulation of FOXO activity is the phosphatidylinositol-3-kinase (PI3K)-AKT signalling pathway, enabling cell survival and proliferation. FOXO family members can be further regulated by distinct kinases, both positively (e.g., JNK, AMPK) and negatively (e.g., ERK-MAPK, CDK2), with additional post-translational modifications further impacting on FOXO activity. Evidence has suggested that FOXOs behave as 'bona fide' tumour suppressors, through transcriptional programmes regulating several cellular behaviours including cell cycle arrest and apoptosis. However, an alternative paradigm has emerged which indicates that FOXOs operate as mediators of cellular homeostasis and/or resistance in both 'normal' and pathophysiological scenarios. Distinct FOXO family members fulfil discrete roles during normal B cell maturation and function, and it is now clear that FOXOs are aberrantly expressed and mutated in discrete B-cell malignancies. While active FOXO function is generally associated with disease suppression in chronic lymphocytic leukemia for example, FOXO expression is associated with disease progression in diffuse large B cell lymphoma, an observation also seen in other cancers. The opposing functions of the FOXO family drives the debate about the circumstances in which FOXOs favour or hinder disease progression, and whether targeting FOXO-mediated processes would be effective in the treatment of B-cell malignancies. Here, we discuss the disparate roles of FOXO family members in B lineage cells, the regulatory events that influence FOXO function focusing mainly on post-translational modifications, and consider the potential for future development of therapies that target FOXO activity.
Collapse
Affiliation(s)
| | | | | | - Alison M. Michie
- Paul O’Gorman Leukaemia Research Centre, School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
2
|
Nguyen-Khac F, Bidet A, Daudignon A, Lafage-Pochitaloff M, Ameye G, Bilhou-Nabéra C, Chapiro E, Collonge-Rame MA, Cuccuini W, Douet-Guilbert N, Eclache V, Luquet I, Michaux L, Nadal N, Penther D, Quilichini B, Terre C, Lefebvre C, Troadec MB, Véronèse L. The complex karyotype in hematological malignancies: a comprehensive overview by the Francophone Group of Hematological Cytogenetics (GFCH). Leukemia 2022; 36:1451-1466. [DOI: 10.1038/s41375-022-01561-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/24/2022] [Accepted: 03/28/2022] [Indexed: 12/16/2022]
|
3
|
Shanmugasundaram K, Goyal S, Switchenko J, Calzada O, Churnetski MC, Kolla B, Bachanova V, Gerson JN, Barta SK, Gordon MJ, Danilov AV, Grover NS, Mathews S, Burkart M, Karmali R, Sawalha Y, Hill BT, Ghosh N, Park SI, Epperla N, Bond DA, Badar T, Blum KA, Hamadani M, Fenske TS, Malecek M, Kahl BS, Martin P, Guo J, Flowers CR, Cohen JB. Intensive induction regimens after deferring initial therapy for mantle cell lymphoma are not associated with improved survival. Eur J Haematol 2021; 107:301-310. [PMID: 33973276 DOI: 10.1111/ejh.13649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 01/05/2023]
Abstract
INTRODUCTION While most patients with mantle cell lymphoma (MCL) receive therapy shortly after diagnosis, a subset of patients with indolent-behaving disease can safely defer treatment. In this subgroup, we evaluated the importance of treatment intensity in patients with MCL who defer initial therapy. METHODS Out of 1134 patients with MCL from 12 academic centers, we analyzed 219 patients who initiated therapy at least 90 days after diagnosis. Patients who received induction with high-dose cytarabine and/or autologous stem cell transplantation (ASCT) in first remission were considered to have received intensive therapy (n = 88) while all other approaches were non-intensive (n = 131). RESULTS There was no difference in progression-free (PFS; P = .224) or overall survival (OS; P = .167) in deferred patients who received non-intensive vs. intensive therapy. Additionally, univariate and multivariate Cox proportional hazards models were performed for PFS and OS. Treatment at an academic center (HR 0.43, P = .015) was associated with improved OS in both univariate and multivariate models, while intensity of treatment was not associated with improved OS in either model. CONCLUSIONS These results indicate that intensified initial treatment is not associated with improved survival after deferring initial therapy, although prospective studies are needed to determine which of these patients with MCL may benefit from intensive therapy.
Collapse
Affiliation(s)
| | - Subir Goyal
- Winship Cancer Institute, Biostatistics and Bioinformatics, Emory University, Atlanta, GA, US
| | - Jeffery Switchenko
- Winship Cancer Institute, Biostatistics and Bioinformatics, Emory University, Atlanta, GA, US
| | - Oscar Calzada
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| | - Michael C Churnetski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| | - Bhaskar Kolla
- Division of Hematology Oncology and Transplantation, University of Minnesota, Minneapolis, MN, US
| | - Veronika Bachanova
- Division of Hematology Oncology and Transplantation, University of Minnesota, Minneapolis, MN, US
| | - James N Gerson
- Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, US
| | - Stefan K Barta
- Hematology and Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, US
| | - Max J Gordon
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, US
| | - Alexey V Danilov
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, US
| | - Natalie S Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, US
| | - Stephanie Mathews
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, US
| | - Madelyn Burkart
- Division of Hematology, Northwestern University, Chicago, IL, US
| | - Reem Karmali
- Division of Hematology, Northwestern University, Chicago, IL, US
| | - Yazeed Sawalha
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, US
| | - Brian T Hill
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, US
| | | | | | - Narendranath Epperla
- Division of Hematology, The Ohio State University Cancer Center, Columbus, OH, US.,Mayo Clinic, Jacksonville, FL, US
| | - David A Bond
- Division of Hematology, The Ohio State University Cancer Center, Columbus, OH, US
| | | | - Kristie A Blum
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| | - Mehdi Hamadani
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, US
| | - Timothy S Fenske
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, US
| | - Mary Malecek
- Siteman Cancer Center, Washington University, St. Louis, MO, US
| | - Brad S Kahl
- Siteman Cancer Center, Washington University, St. Louis, MO, US
| | | | - Jin Guo
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Christopher R Flowers
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University Hospital, Atlanta, GA, US
| |
Collapse
|
4
|
Condoluci A, Rossi D, Zucca E, Cavalli F. Toward a Risk-Tailored Therapeutic Policy in Mantle Cell Lymphoma. Curr Oncol Rep 2018; 20:79. [PMID: 30132080 DOI: 10.1007/s11912-018-0728-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Mantle cell lymphoma (MCL) prognosis is strictly related to the characteristics of the disease, which can range from very indolent cases to highly aggressive and refractory ones. Here we will review the current knowledge on MCL biomarkers. RECENT FINDINGS Biomarker-informed diagnosis is essential for differentiating MCL from other mature B cell tumors. Diagnosis of MCL relies on the identification of the t(11;14) translocation by FISH or the consequently aberrant expression of cyclin D1 by immunohistochemistry. For the few cases staining negative for cyclin D1, SOX11 may help to define the diagnosis. Prognostic biomarkers have been proposed to stratify MCL patients, including baseline clinical aspects (leukemic non-nodal presentation, in situ presentation, Mantle cell International Prognostic Index-MIPI), pathological aspects (blastoid morphology, Ki-67 proliferation index, SOX11 expression), genetic aspects (immunoglobulin gene mutation status, TP53 deletion or mutation, CDKN2A deletion), and depth of response after treatment (PET imaging, molecular minimal residual disease). Such tools are increasingly used as a guide for therapeutic decisions. Watchful waiting approach is recommended for patients harboring favorable clinico-biological features, such as leukemic non-nodal presentation, low MIPI score, non-blastoid disease, low Ki-67 proliferation rate, mutated immunoglobulin genes, and the lack of SOX11 expression. For patients in need of frontline therapy, the decision of whether to undertake intensive regimens is based upon patient's age and comorbidities. Central nervous system prophylaxis is recommended for cases showing blastoid morphology. The duration of remission is tightly correlated to the depth of response. With the aim of achieving a longer duration of remission and survival, younger patients may pursue more intensive regimens incorporating high-dose cytarabine, followed by myeloablative consolidation chemotherapy, autologous stem cell transplantation, and rituximab maintenance. Older patients could, on the other hand, benefit from lower intensity immunochemotherapy followed or not by a maintenance therapy depending on which frontline regimen is used. Despite the identification of several potential useful biomarkers that may inform the treatment decisions and the design of clinical trials, the treatment choice remains nowadays determined by the patient age and fitness rather than by the individual patient characteristics. Tailoring therapy toward a risk-adapted strategy to accommodate the wide spectrum of disease is an urgent challenge, and clinical trials may explore the feasibility of a biomarker-defined therapeutic policy.
Collapse
Affiliation(s)
- Adalgisa Condoluci
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland
| | - Davide Rossi
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland
| | - Emanuele Zucca
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland
| | - Franco Cavalli
- Institute of Oncology Research (IOR), Via Vela 6, 6500, Bellinzona, Switzerland.
| |
Collapse
|
5
|
McKay P, Leach M, Jackson B, Robinson S, Rule S. A British Society for haematology good practice paper on the diagnosis and investigation of patients with mantle cell lymphoma. Br J Haematol 2018; 182:63-70. [PMID: 29882587 DOI: 10.1111/bjh.15281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Pamela McKay
- Department of Haematology; Beatson West of Scotland Cancer Centre; Gartnavel Hospital; Glasgow UK
| | - Mike Leach
- Department of Haematology; Beatson West of Scotland Cancer Centre; Gartnavel Hospital; Glasgow UK
| | - Bob Jackson
- Department of Pathology; Queen Elizabeth University Hospital; Glasgow UK
| | - Stephen Robinson
- Department of Haematology; University Hospitals Bristol; Bristol UK
| | - Simon Rule
- Department of Haematology; Plymouth University Peninsula Schools of Medicine and Dentistry; Plymouth UK
| |
Collapse
|
6
|
Greenwell IB, Staton AD, Lee MJ, Switchenko JM, Saxe DF, Maly JJ, Blum KA, Grover NS, Mathews SP, Gordon MJ, Danilov AV, Epperla N, Fenske TS, Hamadani M, Park SI, Flowers CR, Cohen JB. Complex karyotype in patients with mantle cell lymphoma predicts inferior survival and poor response to intensive induction therapy. Cancer 2018; 124:2306-2315. [PMID: 29579328 DOI: 10.1002/cncr.31328] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/16/2018] [Accepted: 02/05/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND Risk stratification of newly diagnosed patients with mantle cell lymphoma (MCL) primarily is based on the MCL International Prognostic Index (MIPI) and Ki-67 proliferative index. Single-center studies have reported inferior outcomes in patients with a complex karyotype (CK), but this remains an area of controversy. METHODS The authors retrospectively reviewed 483 patients from 5 academic centers in the United States and described the effect of a CK on survival outcomes in individuals with MCL. RESULTS A CK was found to be associated with inferior overall survival (OS) (4.5 vs 11.6 years; P<.01) and progression-free survival (PFS) (1.9 vs 4.4 years; P<.01). In patients who underwent high-intensity induction followed by autologous stem cell transplantation (ASCT) in first remission, a CK was associated with poor OS (5.1 vs 11.6 years; P = .04) and PFS (3.6 vs 7.8 years; P<.01). Among patients with a CK, high-intensity induction had no effect on OS (4.5 vs 3.8 years; P = .77) nor PFS (2.3 vs 1.5 years; P = .46). Similarly, ASCT in first remission did not improve PFS (3.5 vs 1.2 years; P = .12) nor OS (5.1 vs 4.0 years; P = .27). On multivariable analyses with Ki-67 and MIPI, only CK was found to be predictive of OS (hazard ratio [HR], 1.98; 95% confidence interval [95% CI], 1.12-3.49 [P = .02]), whereas both CK (HR, 1.91; 95% CI, 1.17-3.12 [P = .01]) and Ki-67 >30% (HR, 1.86; 95% CI, 1.06-3.28 [P = .03]) were associated with inferior PFS. Multivariable analysis did not identify any specific cytogenetic abnormalities associated with inferior survival. CONCLUSIONS CK appears to be independently associated with inferior outcomes in patients with MCL regardless of the intensity of induction therapy and receipt of ASCT. Cytogenetics should be incorporated into the workup of a new diagnosis of MCL and novel therapeutic approaches should be investigated for patients with CK. Cancer 2018;124:2306-15. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- I Brian Greenwell
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Ashley D Staton
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Michael J Lee
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jeffrey M Switchenko
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Debra F Saxe
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Joseph J Maly
- Division of Hematology/Oncology, The Ohio State University James Cancer Hospital, Columbus, Ohio
| | - Kristie A Blum
- Division of Hematology/Oncology, The Ohio State University James Cancer Hospital, Columbus, Ohio
| | - Natalie S Grover
- Division of Hematology/Oncology, University of North Carolina at Chapel Hill Lineberger Cancer Center, Chapel Hill, North Carolina
| | - Stephanie P Mathews
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill Lineberger Cancer Center, Chapel Hill, North Carolina
| | - Max J Gordon
- Department of Medicine, OHSU Knight Cancer Institute, Portland, Oregon
| | - Alexey V Danilov
- Department of Medicine, OHSU Knight Cancer Institute, Portland, Oregon
| | - Narendranath Epperla
- Department of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | - Timothy S Fenske
- Department of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | - Mehdi Hamadani
- Department of Hematology and Oncology, Medical College of Wisconsin Cancer Center, Milwaukee, Wisconsin
| | | | - Christopher R Flowers
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Jonathon B Cohen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
7
|
Cohen JB, Zain JM, Kahl BS. Current Approaches to Mantle Cell Lymphoma: Diagnosis, Prognosis, and Therapies. Am Soc Clin Oncol Educ Book 2017; 37:512-525. [PMID: 28561694 DOI: 10.1200/edbk_175448] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mantle cell lymphoma (MCL) is a unique lymphoma subtype, both biologically and clinically. Virtually all cases are characterized by a common genetic lesion, t(11;14), resulting in overexpression of cyclin D1. The clinical course is moderately aggressive, and the disease is considered incurable. Considerable biologic and clinical heterogeneity exists, with some patients experiencing a rapidly progressive course, while others have disease that is readily managed. New tools exist for risk stratification and may allow for a more personalized approach in the future. Landmark studies have been completed in recent years and outcomes appear to be improving. Randomized clinical trials have clarified the role of high-dose cytarabine (Ara-C) for younger patients and have demonstrated a role for maintenance rituximab therapy. Multiple areas of uncertainty remain, however, and are the focus of ongoing research. This review focuses on (1) strategies to differentiate between aggressive and less aggressive cases, (2) understanding who should receive hematopoietic stem cell transplantation, and (3) the role for maintenance therapy in MCL.
Collapse
Affiliation(s)
- Jonathon B Cohen
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| | - Jasmine M Zain
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| | - Brad S Kahl
- From Emory University, Atlanta, GA; City of Hope, Duarte, CA; Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
8
|
Gallo M, Cacheux V, Vincent L, Bret C, Tempier A, Guittard C, Macé A, Leventoux N, Costes V, Szablewski V. Leukemic non-nodal mantle cell lymphomas have a distinct phenotype and are associated with deletion of PARP1 and 13q14. Virchows Arch 2016; 469:697-706. [PMID: 27605053 DOI: 10.1007/s00428-016-2016-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/22/2016] [Accepted: 08/30/2016] [Indexed: 11/26/2022]
Abstract
Leukemic non-nodal mantle cell lymphoma (lMCL) is a particular subtype of mantle cell lymphoma (MCL), characterized by leukemic non-nodal disease and slow progression. Recognition of this entity is relevant to avoid overtreatment. Despite indolent clinical behaviour, lMCL might transform to a more aggressive disease. The purpose of this study was to compare lMCL with classical MCL (cMCL) and aggressive MCL (aMCL) using immunohistochemistry, interphase fluorescence in situ hybridization (FISH), and array-based comparative genomic hybridization, in order to identify biomarkers for lMCL diagnosis and prognosis. Seven lMCL patients were included. All had bone marrow involvement without lymphadenopathy. An lMCL phenotype was distinct from that of cMCL and aMCL: SOX11-, ATM+, PARP1+/-, and low KI67 (average 2 %). Beyond the t(11;14) translocation, fewer secondary cytogenetic alterations were found in lMCL compared to cMCL and aMCL, including deletion of PARP1 and 13q14. At last follow-up, one patient with lMCL had died of disease and another had progressive disease. These patients were respectively 13q14 deletion- and PARP1-positive. One other case of lMCL harbored a 13q14 deletion associated with PARP1 deletion. This patient had indolent disease. lMCL has a particular phenotype and fewer secondary cytogenetic alterations than cMCL and aMCL. PARP1 protein expression and 13q14 deletion are associated with a progressive clinical course of lMCL and should be included in initial diagnostic studies as predictors of unfavorable outcome. PARP1 deletion is involved in lMCL pathogenesis and might confer advantage.
Collapse
Affiliation(s)
- Mathieu Gallo
- Département de Biopathologie, Hôpital Gui De Chauliac, CHU Montpellier, 34275, Montpellier, France
- Faculté de Médecine, Université Montpellier, 2 rue école de Médecine, 34060, Montpellier, France
| | - Valère Cacheux
- Département d'Hématologie biologique, Hôpital Saint Eloi, CHU Montpellier, 34275, Montpellier, France
- Faculté de Médecine, Université Montpellier, 2 rue école de Médecine, 34060, Montpellier, France
| | - Laure Vincent
- Département d'Hématologie clinique, Hôpital Saint Eloi, CHU Montpellier, 34275, Montpellier, France
- Faculté de Médecine, Université Montpellier, 2 rue école de Médecine, 34060, Montpellier, France
| | - Caroline Bret
- Département d'Hématologie biologique, Hôpital Saint Eloi, CHU Montpellier, 34275, Montpellier, France
- Faculté de Médecine, Université Montpellier, 2 rue école de Médecine, 34060, Montpellier, France
| | - Ariane Tempier
- Département de Biopathologie, Hôpital Gui De Chauliac, CHU Montpellier, 34275, Montpellier, France
| | - Caroline Guittard
- Département d'Hématologie biologique, Hôpital Saint Eloi, CHU Montpellier, 34275, Montpellier, France
| | - Alexandra Macé
- Département d'Hématologie biologique, Hôpital Saint Eloi, CHU Montpellier, 34275, Montpellier, France
| | - Nicolas Leventoux
- Département de Biopathologie, Hôpital Gui De Chauliac, CHU Montpellier, 34275, Montpellier, France
| | - Valérie Costes
- Département de Biopathologie, Hôpital Gui De Chauliac, CHU Montpellier, 34275, Montpellier, France
- Faculté de Médecine, Université Montpellier, 2 rue école de Médecine, 34060, Montpellier, France
| | - Vanessa Szablewski
- Département de Biopathologie, Hôpital Gui De Chauliac, CHU Montpellier, 34275, Montpellier, France.
- Faculté de Médecine, Université Montpellier, 2 rue école de Médecine, 34060, Montpellier, France.
- Département de Biopathologie Cellulaire et Tissulaire des Tumeurs, Hôpital Gui De Chauliac, CHU Montpellier, 34275, Montpellier, France.
| |
Collapse
|
9
|
Peluso AL, Ieni A, Mignogna C, Zeppa P. Lymph Node Fine-Needle Cytology: Beyond Flow Cytometry. Acta Cytol 2016; 60:372-384. [PMID: 27560152 DOI: 10.1159/000447734] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022]
Abstract
Lymph node (LN) fine-needle cytology (FNC) coupled with flow cytometry immunophenotyping provides relevant information for the diagnosis of non-Hodgkin lymphoma (NHL). Numerous studies have shown FNC samples to be suitable for different molecular procedures; in this review, some of the molecular procedures most commonly employed for NHL are briefly described and evaluated in this perspective. Fluorescence in situ hybridization and chromogenic in situ hybridization are briefly described. Polymerase chain reaction (PCR)-based assays are used to identify and quantify mutations and translocations, namely immunoglobulin (IGH) and T-cell receptor rearrangements by clonality testing and IGVH somatic hypermutations either by Sanger sequencing, single-strand conformational polymorphisms or RT-PCR strategies. High-throughput technologies (HTT) encompass numerous and different diagnostic tools that share the capacity of multiple molecular investigation and sample processing in a fast and reproducible manner. HTT includes gene expression profiling, comparative genomic hybridization, single-nucleotide polymorphism arrays and next-generation sequencing technologies. A brief description of these tools and their potential application to LN FNC is reported. The challenge for FNC will be to achieve new knowledge and apply new technologies to FNC, exploiting its own basic qualities.
Collapse
Affiliation(s)
- Anna Lucia Peluso
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | | | | | | |
Collapse
|
10
|
Hsi AC, Hurley MY, Lee SJ, Rosman IS, Pang X, Gru A, Schaffer A. Diagnostic utility of SOX11 immunohistochemistry in differentiating cutaneous spread of mantle cell lymphoma from primary cutaneous B-cell lymphomas. J Cutan Pathol 2016; 43:354-61. [PMID: 26762898 DOI: 10.1111/cup.12668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 01/10/2016] [Accepted: 01/10/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mantle cell lymphoma (MCL) is associated with the worst prognosis among low-grade B-cell lymphomas. While cutaneous involvement by nodal or systemic MCL is uncommon, its differentiation from primary cutaneous B-cell lymphoma (CBCL) or cutaneous involvement by other extra-cutaneous BCL is challenging as neither histomorphology nor immunophenotype can be absolutely specific. We analyzed the diagnostic utility of SOX11 immunohistochemistry in differentiating secondary cutaneous MCL from other low-grade CBCL. METHODS Immunohistochemical staining with anti-SOX11 antibody was performed on 8 cases of secondary cutaneous MCL, 16 secondary cutaneous CLL, 20 primary cutaneous MZL, 12 cutaneous FCL (6 primary, 6 secondary), 7 primary cutaneous DLBCL, leg type, 5 systemic DLBCL and 3 B-ALL. SOX11 and cyclin D1 staining were compared in secondary cutaneous MCL. RESULTS Nuclear SOX11 staining was seen in seven of eight cases (88%) of secondary cutaneous MCL, including a case with minimal cyclin D1 expression. All other CBCL lacked detectable nuclear SOX11 expression. The sensitivity and specificity for SOX11 in MCL were 87.5 and 100%, respectively. Both the sensitivity and specificity for combined SOX11 and cyclin D1 immunohistochemistry were 100%. CONCLUSION SOX11 immunohistochemistry could be a useful adjunct in distinguishing secondary cutaneous MCL from other CBCL.
Collapse
Affiliation(s)
- Andy C Hsi
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - M Yadira Hurley
- Department of Dermatology, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Sena J Lee
- Division of Dermatology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ilana S Rosman
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.,Division of Dermatology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Xiaofan Pang
- Department of Biology, Washington University in St. Louis, Saint Louis, MO, USA
| | - Alejandro Gru
- Department of Pathology, The Ohio State University Medical Center, Columbus, OH, USA
| | - András Schaffer
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.,Division of Dermatology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
11
|
Mantle cell lymphoma—a spectrum from indolent to aggressive disease. Virchows Arch 2015; 468:245-57. [DOI: 10.1007/s00428-015-1840-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/17/2015] [Indexed: 01/18/2023]
|
12
|
High-dose cytarabine does not overcome the adverse prognostic value of CDKN2A and TP53 deletions in mantle cell lymphoma. Blood 2015; 126:604-11. [DOI: 10.1182/blood-2015-02-628792] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/24/2015] [Indexed: 12/25/2022] Open
Abstract
Key Points
CDKN2A and TP53 deletions remain of bad prognostic value in younger MCL patients treated according to the current standard of care. CDKN2A and TP53 deletions have independent deleterious effects and should be considered for treatment decisions in addition to MIPI and Ki-67 index.
Collapse
|
13
|
Cohen JB, Ruppert AS, Heerema NA, Andritsos LA, Jones JA, Porcu P, Baiocchi R, Christian BA, Byrd JC, Flynn J, Penza S, Devine SM, Blum KA. Complex Karyotype Is Associated With Aggressive Disease and Shortened Progression-Free Survival in Patients With Newly Diagnosed Mantle Cell Lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2015; 15:278-285.e1. [DOI: 10.1016/j.clml.2014.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 10/24/2022]
|
14
|
Ozer O, Toprak SK, Ote E, Yılmaz Z, Sahin FI. Blastoid variant mantle cell lymphoma with complex karyotype including 11q duplication. Turk J Haematol 2014; 31:290-4. [PMID: 25330523 PMCID: PMC4287031 DOI: 10.4274/tjh.2012.0195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
We describe a case of blastoid mantle cell lymphoma with a complex karyotype. The blastoid variant is a rare type of non-Hodgkin lymphoma exhibiting an aggressive clinical course. Mantle cell lymphoma is a distinct entity of mature B-cell neoplasms genetically characterized by the presence of t(11;14). In the present case, conventional analysis revealed structural abnormalities of chromosomes 2, 4, 6, 10, 13, and 19, along with 3 additional marker chromosomes. The derivative 1 chromosome determined in the case was a result of t(1p;11q). Our interesting finding was the presence of a different translocation between 11q and chromosome 1 in addition to t(11;14). Thus, the resulting 11q duplication was believed to additionally increase the enhanced expression of cyclin D1 gene, which is responsible in the pathogenesis of the disease. Fluorescence in situ hybridization method by the t(11;14) probe revealed clonal numerical abnormalities of chromosomes 11 and 14 in some cells. The detection of multiple abnormalities explains the bad prognosis in the present case. On the basis of our findings, we can easily conclude that results of cytogenetic analyses of similar mantle cell lymphoma patients would provide clues about new responsible gene regions and disease prognosis. In conclusion, it has been suggested that the presence of multiple chromosomal aberrations in addition to the specific t(11;14) may have a negative impact on clinical course and survival rate.
Collapse
Affiliation(s)
- Ozge Ozer
- Başkent University Faculty of Medicine, Department of Hematology, Ankara, Turkey. E-ma-il:
| | | | | | | | | |
Collapse
|
15
|
Wang M, Popplewell LL, Collins RH, Winter JN, Goy A, Kaminski MS, Bartlett NL, Johnston PB, Lister J, Fanning SR, Tuscano JM, Beck JT, Kaya H, Robeva A, Fan J, Klimovsky J, Cheung W, Cherfi A, O'Connor OA. Everolimus for patients with mantle cell lymphoma refractory to or intolerant of bortezomib: multicentre, single-arm, phase 2 study. Br J Haematol 2014; 165:510-8. [PMID: 24579926 DOI: 10.1111/bjh.12780] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/06/2013] [Indexed: 02/03/2023]
Abstract
The multicentre, open-label, two-stage, single-arm, phase 2, PILLAR (PIvotaL Lymphoma triAls of RAD001)-1 study (NCT00702052) assessed the efficacy and safety of everolimus 10 mg/d in adults with confirmed mantle cell lymphoma (MCL) refractory to or intolerant of bortezomib who received ≥1 other antineoplastic agent, either separately or in combination with bortezomib. Primary endpoint was overall response rate (ORR) per investigator review according to the response criteria for malignant lymphoma. Secondary endpoints included progression-free survival (PFS), overall survival (OS) and safety. Fifty-eight patients were enrolled from August 2008-January 2011. Five partial responses were observed (ORR 8·6%; 90% confidence interval [CI] 3·5-17·3%); the study did not meet the prespecified objective of ≥8 objective responses among 57 patients. Median PFS and OS were 4·4 months (95% CI 3·5-6·1) and 16·9 months (95% CI 14·4-29·9), respectively. Grade 3/4 non-haematological toxicities occurred in 70·7% of patients. Based on laboratory values, grade 3/4 thrombocytopenia, neutropenia and anaemia occurred in 13·8%, 13·8% and 8·6% of patients, respectively. Everolimus demonstrated modest activity and acceptable tolerability in heavily pretreated patients with MCL refractory to or intolerant of bortezomib. Future studies evaluating everolimus in a less refractory population or in combination with other targeted therapies in refractory MCL are warranted.
Collapse
Affiliation(s)
- Michael Wang
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Lu TX, Li JY, Xu W. The role of SOX11 in mantle cell lymphoma. Leuk Res 2013; 37:1412-9. [DOI: 10.1016/j.leukres.2013.07.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 07/26/2013] [Accepted: 07/27/2013] [Indexed: 12/14/2022]
|
17
|
Sarkozy C, Terré C, Jardin F, Radford I, Roche-Lestienne C, Penther D, Bastard C, Rigaudeau S, Pilorge S, Morschhauser F, Bouscary D, Delarue R, Farhat H, Rousselot P, Hermine O, Tilly H, Chevret S, Castaigne S. Complex karyotype in mantle cell lymphoma is a strong prognostic factor for the time to treatment and overall survival, independent of the MCL international prognostic index. Genes Chromosomes Cancer 2013; 53:106-16. [DOI: 10.1002/gcc.22123] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 10/09/2013] [Indexed: 12/22/2022] Open
Affiliation(s)
- Clémentine Sarkozy
- Department of Hematology; Centre Hospitalier de Versailles, Le Chesnay; Université de Versailles-Saint Quentin; Versailles France
| | - Christine Terré
- Department of Cytogenetics; Centre Hospitalier de Versailles; Le Chesnay France
| | - Fabrice Jardin
- Department of Hematology and INSERM U918; Centre Henri Becquerel; Rouen France
| | - Isabelle Radford
- Department of Cytogenetics; Necker Hospital, AP-HP; Paris France
| | | | - Dominique Penther
- Department of Cytogenetics; Centre Hospitalier Henri Becquerel; Rouen France
| | - Christian Bastard
- Department of Cytogenetics; Centre Hospitalier Henri Becquerel; Rouen France
| | - Sophie Rigaudeau
- Department of Hematology; Centre Hospitalier de Versailles, Le Chesnay; Université de Versailles-Saint Quentin; Versailles France
| | - Sylvain Pilorge
- Department of Hematology; Centre Hospitalier de Versailles, Le Chesnay; Université de Versailles-Saint Quentin; Versailles France
| | | | - Didier Bouscary
- Department of Hematology; Cochin Hospital, AP-HP; Paris France
| | - Richard Delarue
- Department of Hematology; Necker Hospital, AP-HP; Paris France
| | - Hassan Farhat
- Department of Hematology; Centre Hospitalier de Versailles, Le Chesnay; Université de Versailles-Saint Quentin; Versailles France
| | - Philippe Rousselot
- Department of Hematology; Centre Hospitalier de Versailles, Le Chesnay; Université de Versailles-Saint Quentin; Versailles France
| | - Olivier Hermine
- Department of Hematology; Necker Hospital, AP-HP; Paris France
| | - Hervé Tilly
- Department of Hematology and INSERM U918; Centre Henri Becquerel; Rouen France
| | - Sylvie Chevret
- Department of Biostatistics; Saint Louis Hospital, AP-HP; Université Paris-Diderot; Inserm S717 Paris France
| | - Sylvie Castaigne
- Department of Hematology; Centre Hospitalier de Versailles, Le Chesnay; Université de Versailles-Saint Quentin; Versailles France
| |
Collapse
|
18
|
Fang C, Dong HJ, Zou ZJ, Fan L, Wang L, Zhang R, Xu J, Xu W, Li JY. High expression of cyclic nucleotide phosphodiesterase 7B mRNA predicts poor prognosis in mantle cell lymphoma. Leuk Res 2013; 37:536-40. [DOI: 10.1016/j.leukres.2013.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 01/30/2013] [Accepted: 02/05/2013] [Indexed: 12/20/2022]
|
19
|
Liu F, Yoshida N, Suguro M, Kato H, Karube K, Arita K, Yamamoto K, Tsuzuki S, Oshima K, Seto M. Clonal heterogeneity of mantle cell lymphoma revealed by array comparative genomic hybridization. Eur J Haematol 2013; 90:51-8. [PMID: 23110670 DOI: 10.1111/ejh.12030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2012] [Indexed: 12/28/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B-cell non-Hodgkin lymphoma (NHL) characterized by the translocation t(11;14)(q13;q32). This lymphoma exhibits a poor prognosis and remains incurable with standard chemotherapy approaches. Recently, we have shown that a majority of patients with acute-type adult T-cell leukemia/lymphoma (ATLL) have multiple subclones that were likely produced in lymph nodes. We investigated whether MCL has multiple subclones as identified in ATLL by high-resolution oligo-array comparative genomic hybridization (CGH). Eleven of 20 (55%) evaluable MCL cases had a log2 ratio imbalance, suggesting the existence of multiple subclones in MCL. Based on the proportion of every subclone relative to the main clone, we were able to speculate clonal evolution in each MCL case with multiple subclones. Our analysis gave new insights into the clonal heterogeneity quantitatively and accurately. Furthermore, genomic copy number alterations are not hierarchical events and not necessarily the initial or later events for cells to become MCL.
Collapse
Affiliation(s)
- Fang Liu
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Genomic profiling of mantle cell lymphoma (MCL) cells has enabled a better understanding of the complex mechanisms underlying the pathogenesis of disease. Besides the t(11;14)(q13;q32) leading to cyclin D1 overexpression, MCL exhibits a characteristic pattern of DNA copy number aberrations that differs from those detected in other B-cell lymphomas. These genomic changes disrupt selected oncogenes and suppressor genes that are required for lymphoma development and progression, many of which are components of cell cycle, DNA damage response and repair, apoptosis, and cell-signaling pathways. Additionally, some of them may represent effective therapeutic targets. A number of genomic and molecular abnormalities have been correlated with the clinical outcome of patients with MCL and are considered prognostic factors. However, only a few genomic markers have been shown to predict the response to current or novel targeted therapies. One representative example is the high-level amplification of the BCL2 gene, which predicts a good response to pro-apoptotic BH3 mimetic drugs. In summary, genomic analyses have contributed to the substantial advances made in the comprehension of the pathogenesis of MCL, providing a solid basis for the identification of optimal therapeutic targets and for the design of new molecular therapies aiming to cure this fatal disease.
Collapse
Affiliation(s)
- Melissa Rieger Menanteau
- Division of Oncology, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | |
Collapse
|
21
|
McKay P, Leach M, Jackson R, Cook G, Rule S. Guidelines for the investigation and management of mantle cell lymphoma. Br J Haematol 2012; 159:405-26. [PMID: 22994971 DOI: 10.1111/bjh.12046] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- P McKay
- Department of Haematology, Beatson West of Scotland Cancer Centre, Gartnavel Hospital, Glasgow, UK
| | | | | | | | | |
Collapse
|
22
|
Abstract
Abstract
The prognostic role of the transcription factor SOX11 in mantle cell lymphoma (MCL) is controversial. We investigated prognostic markers in a population-based cohort of 186 MCL cases. Seventeen patients (9%) did not require any therapy within the first 2 years after diagnosis and were retrospectively defined as having an indolent disease. As expected, indolent MCL had less frequent B symptoms and extensive nodal involvement and 88% of these cases expressed SOX11. In our cohort 13 cases (7.5%) lacked nuclear SOX11 at diagnosis. SOX11− MCL had a higher frequency of lymphocytosis, elevated level of lactate dehydrogenase (LDH), and p53 positivity. The overall survival in the whole cohort, excluding 37 patients receiving autologous stem cell transplantation, was 3.1 year and in patients with indolent or nonindolent disease, 5.9 and 2.8 years, respectively (P = .004). SOX11− cases had a shorter overall survival, compared with SOX11+ cases, 1.5 and 3.2 years, respectively (P = .014). In multivariate analysis of overall survival, age > 65 (P = .001), Eastern Cooperative Oncology Group score ≥ 2 (P = .022), elevated LDH level (P = .001), and p53 expression (P = .001) remained significant, and SOX11 lost significance. We conclude that most indolent MCLs are SOX11+ and that SOX11 cannot be used for predicting an indolent disease course.
Collapse
|
23
|
TP53 mutation is not an independent prognostic factor in patients with mantle cell lymphoma at advanced stage. Med Oncol 2011; 29:2166-73. [DOI: 10.1007/s12032-011-0096-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 10/18/2011] [Indexed: 10/15/2022]
|
24
|
Royo C, Salaverria I, Hartmann EM, Rosenwald A, Campo E, Beà S. The complex landscape of genetic alterations in mantle cell lymphoma. Semin Cancer Biol 2011; 21:322-34. [DOI: 10.1016/j.semcancer.2011.09.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 09/12/2011] [Indexed: 11/29/2022]
|
25
|
Sander B. Mantle cell lymphoma: recent insights into pathogenesis, clinical variability, and new diagnostic markers. Semin Diagn Pathol 2011; 28:245-55. [DOI: 10.1053/j.semdp.2011.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
26
|
|
27
|
da Cunha Santos G, Ko HM, Geddie WR, Boerner SL, Lai SW, Have C, Kamel-Reid S, Bailey D. Targeted use of fluorescence in situ hybridization (FISH) in cytospin preparations. Cancer Cytopathol 2010; 118:250-8. [DOI: 10.1002/cncy.20098] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
28
|
Espinet B, Salaverria I, Beà S, Ruiz-Xivillé N, Balagué O, Salido M, Costa D, Carreras J, Rodríguez-Vicente AE, Luís García J, Hernández-Rivas JM, Calasanz MJ, Siebert R, Ferrer A, Salar A, Carrió A, Polo N, García-Marco JA, Domingo A, González-Barca E, Romagosa V, Marugán I, López-Guillermo A, Millá F, Luís Mate J, Luño E, Sanzo C, Collado R, Oliver I, Monzó S, Palacín A, González T, Sant F, Salinas R, Ardanaz MT, Font L, Escoda L, Florensa L, Serrano S, Campo E, Solé F. Incidence and prognostic impact of secondary cytogenetic aberrations in a series of 145 patients with mantle cell lymphoma. Genes Chromosomes Cancer 2010; 49:439-51. [PMID: 20143418 DOI: 10.1002/gcc.20754] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Mantle cell lymphoma (MCL) is a mature B-cell neoplasm with an aggressive behavior, characterized by the t(11;14)(q13;q32). Several secondary genetic abnormalities with a potential role in the oncogenic process have been described. Studies of large MCL series using conventional cytogenetics, and correlating with proliferation and survival, are scarce. We selected 145 MCL cases at diagnosis, displaying an aberrant karyotype, from centers belonging to the Spanish Cooperative Group for Hematological Cytogenetics. Histological subtype, proliferative index and survival data were ascertained. Combined cytogenetic and molecular analyses detected CCND1 translocations in all cases, mostly t(11;14)(q13;q32). Secondary aberrations were present in 58% of patients, the most frequent being deletions of 1p, 13q and 17p, 10p alterations and 3q gains. The most recurrent breakpoints were identified at 1p31-32, 1p21-22, 17p13, and 1p36. Aggressive blastoid/pleomorphic variants displayed a higher karyotypic complexity, a higher frequency of 1p and 17p deletions and 10p alterations, a higher proliferation index and poor survival. Gains of 3q and 13q and 17p13 losses were associated with reduced survival times. Interestingly, gains of 3q and 17p losses added prognostic significance to the morphology in a multivariate analysis. Our findings confirm previous observations indicating that proliferation index, morphology and several secondary genetic alterations (3q gains and 13q and 17p losses) have prognostic value in patients with MCL. Additionally, we observed that 3q gains and 17p losses detected by conventional cytogenetics are proliferation-independent prognostic markers indicating poor outcome.
Collapse
Affiliation(s)
- Blanca Espinet
- Laboratori de Citogenètica Molecular, Laboratori de Citologia Hematològica, Servei de Patologia, Servei d'Hematologia Clínica, GRETNHE, IMIM-Hospital del Mar, Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pathway discovery in mantle cell lymphoma by integrated analysis of high-resolution gene expression and copy number profiling. Blood 2010; 116:953-61. [PMID: 20421449 DOI: 10.1182/blood-2010-01-263806] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The genome of mantle cell lymphoma (MCL) is, in addition to the translocation t(11;14), characterized by a high number of secondary chromosomal gains and losses that probably account for the various survival times of MCL patients. We investigated 77 primary MCL tumors with available clinical information using high-resolution RNA expression and genomic profiling and applied our recently developed gene expression and dosage integrator algorithm to identify novel genes and pathways that may be of relevance for the pathobiology of MCL. We show that copy number neutral loss of heterozygosity is common in MCL and targets regions that are frequently affected by deletions. The molecular consequences of genomic copy number changes appear complex, even in genomic loci with identified tumor suppressors, such as the region 9p21 containing the CDKN2A locus. Moreover, the deregulation of novel genes, such as CUL4A, ING1, and MCPH1, may affect the 2 crucial pathogenetic mechanisms in MCL, the disturbance of the proliferation, and DNA damage response pathways. Deregulation of the Hippo pathway may have a pathogenetic role in MCL because decreased expression of its members MOBKL2A, MOBKL2B, and LATS2 was associated with inferior outcome, including an independent validation series of 32 MCLs.
Collapse
|
30
|
Fernàndez V, Salamero O, Espinet B, Solé F, Royo C, Navarro A, Camacho F, Beà S, Hartmann E, Amador V, Hernández L, Agostinelli C, Sargent RL, Rozman M, Aymerich M, Colomer D, Villamor N, Swerdlow SH, Pileri SA, Bosch F, Piris MA, Montserrat E, Ott G, Rosenwald A, López-Guillermo A, Jares P, Serrano S, Campo E. Genomic and gene expression profiling defines indolent forms of mantle cell lymphoma. Cancer Res 2010; 70:1408-18. [PMID: 20124476 DOI: 10.1158/0008-5472.can-09-3419] [Citation(s) in RCA: 334] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mantle cell lymphoma (MCL) is typically a very aggressive disease with poor outcomes, but some cases display an indolent behavior that might not necessitate treatment at diagnosis. To define molecular criteria that might permit recognition of such cases, we compared the clinicopathologic features, gene expression, and genomic profile of patients who had indolent or conventional disease (iMCL or cMCL). Patients with iMCL displayed nonnodal leukemic disease with predominantly hypermutated IGVH and noncomplex karyotypes. iMCL and cMCL shared a common gene expression profile that differed from other leukemic lymphoid neoplasms. However, we identified a signature of 13 genes that was highly expressed in cMCL but underexpressed in iMCL. SOX11 was notable in this signature and we confirmed a restriction of SOX11 protein expression to cMCL. To validate the potential use of SOX11 as a biomarker for cMCL, we evaluated SOX11 protein expression in an independent series of 112 cases of MCL. Fifteen patients with SOX11-negative tumors exhibited more frequent nonnodal presentation and better survival compared with 97 patients with SOX11-positive MCL (5-year overall survival of 78% versus 36%, respectively; P = 0.001). In conclusion, we defined nonnodal presentation, predominantly hypermutated IGVH, lack of genomic complexity, and absence of SOX11 expression as qualities of a specific subtype of iMCL with excellent outcomes that might be managed more conservatively than cMCL.
Collapse
Affiliation(s)
- Verònica Fernàndez
- Hematopathology Section, Department of Pathology, Hospital Clinic, Institut d'Investigacions Biomediques August Pi i Sunyer and Department of Hematology, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mozos A, Royo C, Hartmann E, De Jong D, Baró C, Valera A, Fu K, Weisenburger DD, Delabie J, Chuang SS, Jaffe ES, Ruiz-Marcellan C, Dave S, Rimsza L, Braziel R, Gascoyne RD, Solé F, López-Guillermo A, Colomer D, Staudt LM, Rosenwald A, Ott G, Jares P, Campo E. SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype. Haematologica 2010; 94:1555-62. [PMID: 19880778 DOI: 10.3324/haematol.2009.010264] [Citation(s) in RCA: 271] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cyclin D1-negative mantle cell lymphoma is difficult to distinguish from other small B-cell lymphomas. The clinical and pathological characteristics of patients with this form of lymphoma have not been well defined. Overexpression of the transcription factor SOX11 has been observed in conventional mantle cell lymphoma. The aim of this study was to determine whether this gene is expressed in cyclin D1-negative mantle cell lymphoma and whether its detection may be useful to identify these tumors. DESIGN AND METHODS The microarray database of 238 mature B-cell neoplasms was re-examined. SOX11 protein expression was investigated immunohistochemically in 12 cases of cyclin D1-negative mantle cell lymphoma, 54 cases of conventional mantle cell lymphoma, and 209 additional lymphoid neoplasms. RESULTS SOX11 mRNA was highly expressed in conventional and cyclin D1-negative mantle cell lymphoma and in 33% of the cases of Burkitt's lymphoma but not in any other mature lymphoid neoplasm. SOX11 nuclear protein was detected in 50 cases (93%) of conventional mantle cell lymphoma and also in the 12 cyclin D1-negative cases of mantle cell lymphoma, the six cases of lymphoblastic lymphomas, in two of eight cases of Burkitt's lymphoma, and in two of three T-prolymphocytic leukemias but was negative in the remaining lymphoid neoplasms. Cyclin D2 and D3 mRNA levels were significantly higher in cyclin D1-negative mantle cell lymphoma than in conventional mantle cell lymphoma but the protein expression was not discriminative. The clinico-pathological features and outcomes of the patients with cyclin D1-negative mantle cell lymphoma identified by SOX11 expression were similar to those of patients with conventional mantle cell lymphoma. CONCLUSIONS SOX11 mRNA and nuclear protein expression is a highly specific marker for both cyclin D1-positive and negative mantle cell lymphoma.
Collapse
Affiliation(s)
- Ana Mozos
- Hematopathology Section, Department of Pathology and Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
New developments in the pathology of malignant lymphoma: a review of the literature published from May to July 2008. J Hematop 2009; 1:145-60. [PMID: 19669214 PMCID: PMC2713479 DOI: 10.1007/s12308-008-0012-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|