Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Theranostics ; 11(8): 3565-3579, 2021.
Article in English | MEDLINE | ID: mdl-33664848

ABSTRACT

Rationale: Primary central nervous system diffuse large B-cell lymphoma (PCNSL) is a rare and aggressive entity that resides in an immune-privileged site. The tumor microenvironment (TME) and the disruption of the immune surveillance influence lymphoma pathogenesis and immunotherapy resistance. Despite growing knowledge on heterogeneous therapeutic responses, no comprehensive description of the PCNSL TME is available. We hence investigated the immune subtypes of PCNSL and their association with molecular signaling and survival. Methods: Analysis of PCNSL transcriptomes (sequencing, n = 20; microarrays, n = 34). Integrated correlation analysis and signaling pathway topology enabled us to infer intercellular interactions. Immunohistopathology and digital imaging were used to validate bioinformatic results. Results: Transcriptomics revealed three immune subtypes: immune-rich, poor, and intermediate. The immune-rich subtype was associated to better survival and characterized by hyper-activation of STAT3 signaling and inflammatory signaling, e.g., IFNγ and TNF-α, resembling the hot subtype described in primary testicular lymphoma and solid cancer. WNT/ß-catenin, HIPPO, and NOTCH signaling were hyper-activated in the immune-poor subtype. HLA down-modulation was clearly associated with a low or intermediate immune infiltration and the absence of T-cell activation. Moreover, HLA class I down-regulation was also correlated with worse survival with implications on immune-intermediate PCNSL that frequently feature reduced HLA expression. A ligand-receptor intercellular network revealed high expression of two immune checkpoints, i.e., CTLA-4/CD86 and TIM-3/LAGLS9. TIM-3 and galectin-9 proteins were clearly upregulated in PCNSL. Conclusion: Altogether, our study reveals that patient stratification according to immune subtypes, HLA status, and immune checkpoint molecule quantification should be considered prior to immune checkpoint inhibitor therapy. Moreover, TIM-3 protein should be considered an axis for future therapeutic development.


Subject(s)
Central Nervous System Neoplasms/immunology , Lymphoma, Large B-Cell, Diffuse/immunology , Adult , Aged , Aged, 80 and over , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Cohort Studies , Down-Regulation , Female , Gene Expression Profiling , HLA Antigens/genetics , Humans , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Precision Medicine , Prognosis , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
J Clin Pathol ; 74(10): 650-656, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32912960

ABSTRACT

AIMS: Diffuse large B-cell lymphoma (DLBCL) is the most common type of aggressive non-Hodgkin's lymphoma that represents a heterogeneous group of disease that is differentially characterised by clinical, molecular and cytogenetic features. MYC, BCL2 and BCL6 gene rearrangements have been identified as prognostic factors in DLBCL, especially for MYC. Nevertheless the frequency and effect of atypical/unbalanced BCL6, BCL2 and MYC translocations in DLBCL is not fully documented. Here, we aimed to analyse those atypical/unbalanced rearrangements in DLBCL and to assess their prognostic impact. METHODS: We collected tumour tissue and clinical data from 97 DLBCL and used interphase fluorescence in situ hybridisation (FISH) with break-apart probe to characterise BCL6, BCL2 and MYC gene pattern. RESULTS: 19 of 97 (19,6%) cases of DLBCL had atypical/ unbalanced gene rearrangements (14 involving BCL6 gene, 5 involving BCL2 gene and none involving MYC gene). Compared with patients with simple gene rearrangement and patients without cytogenetic abnormality, patients with atypical/unbalanced gene rearrangement were in an unfavourable risk group by the International Prognostic Index (p=0039), died of disease (p=0012), harboured relapse or progression (p=0011) and had shorter overall (p=0,04), relapse free (p=0029) and event free (p=0026) survival. CONCLUSIONS: We showed that patients with DLBCL with BCL2 or BCL6 atypical/unbalanced rearrangements constituted a group of patients with poor outcome. We also underlined the importance of FISH analyses, easily feasible in routine practise, at diagnosis of DLBCL to detect the rather frequent and clinically significant atypical/unbalanced aberrations of these genes.


Subject(s)
Biomarkers, Tumor/genetics , Gene Rearrangement , Lymphoma, Large B-Cell, Diffuse/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , In Situ Hybridization, Fluorescence , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Male , Middle Aged , Progression-Free Survival , Recurrence , Young Adult
3.
J Clin Med ; 9(7)2020 Jul 12.
Article in English | MEDLINE | ID: mdl-32664688

ABSTRACT

Donor lymphocyte infusion (DLI) is used to prevent or treat haematological malignancies relapse after allogeneic stem cell transplantation (allo-SCT). Recombinant human granulocyte colony-stimulated factor primed DLI (gDLI) is derived from frozen aliquots of the peripheral blood stem cell collection. We compared the efficacy and safety of gDLI and classical DLI after allo-SCT. We excluded haploidentical allo-SCT. Initial diseases were acute myeloblastic leukaemia (n = 45), myeloma (n = 38), acute lymphoblastic leukaemia (n = 20), non-Hodgkin lymphoma (n = 10), myelodysplasia (n = 8), Hodgkin lymphoma (n = 8), chronic lymphocytic leukaemia (n = 7), chronic myeloid leukaemia (n = 2) and osteomyelofibrosis (n = 1). Indications for DLI were relapse (n = 96) or pre-emptive treatment (n = 43). Sixty-eight patients had classical DLI and 71 had gDLI. The response rate was 38.2%, the 5-year progression-free survival (PFS) rate was 38% (29-48) and the 5-year overall survival (OS) rate was 37% (29-47). Graft versus host disease rate was 46.7% and 10.1% of patients died from toxicity. There were no differences between classical DLI and gDLI in terms of response (p = 0.28), 5-year PFS (p = 0.90), 5-year OS (p. 0.50), GvHD (p = 0.86), treated GvHD (p = 0.81) and cause of mortality (p. 0.14). In conclusion, this study points out no major effectiveness or toxicity of gDLI compared to classical DLI.

4.
Theranostics ; 10(10): 4383-4394, 2020.
Article in English | MEDLINE | ID: mdl-32292502

ABSTRACT

Purpose: Salivary duct carcinoma (SDC) is a rare and aggressive salivary gland cancer subtype with poor prognosis. The mutational landscape of SDC has already been the object of several studies, however little is known regarding the functional genomics and the tumor microenvironment despite their importance in oncology. Our investigation aimed at describing both the functional genomics of SDC and the SDC microenvironment, along with their clinical relevance. Methods: RNA-sequencing (24 tumors), proteomics (17 tumors), immunohistochemistry (22 tumors), and multiplexed immunofluorescence (3 tumors) data were obtained from three different patient cohorts and analyzed by digital imaging and bioinformatics. Adjacent non-tumoral tissue from patients in two cohorts were used in transcriptomic and proteomic analyses. Results: Transcriptomic and proteomic data revealed the importance of Notch, TGF-ß, and interferon-γ signaling for all SDCs. We confirmed an overall strong desmoplastic reaction by measuring α-SMA abundance, the level of which was associated with recurrence-free survival (RFS). Two distinct immune phenotypes were observed: immune-poor SDCs (36%) and immune-infiltrated SDCs (64%). Advanced bioinformatics analysis of the transcriptomic data suggested 72 ligand-receptor interactions occurred in the microenvironment and correlated with the immune phenotype. Among these interactions, three immune checkpoints were validated by immunofluorescence, including CTLA-4/DC86 and TIM-3/galectin-9 interactions, previously unidentified in SDC. Immunofluorescence analysis also confirmed an important immunosuppressive role of macrophages and NK cells, also supported by the transcriptomic data. Conclusions: Together our data significantly increase the understanding of SDC biology and open new perspectives for SDC tumor treatment. Before applying immunotherapy, patient stratification according to the immune infiltrate should be taken into account. Immune-infiltrated SDC could benefit from immune checkpoint-targeting therapy, with novel options such as anti-CTLA-4. Macrophages or NK cells could also be targeted. The dense stroma, i.e., fibroblasts or hyaluronic acid, may also be the focus for immune-poor SDC therapies, e.g. in combination with Notch or TGF-ß inhibitors, or molecules targeting SDC mutations.


Subject(s)
Carcinoma, Ductal , Salivary Ducts/immunology , Salivary Gland Neoplasms , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinoma, Ductal/genetics , Carcinoma, Ductal/immunology , Cohort Studies , Female , Humans , Male , Middle Aged , Proteome , Salivary Ducts/pathology , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/immunology , Transcriptome , Young Adult
5.
Virchows Arch ; 476(6): 891-902, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31811434

ABSTRACT

Primary central nervous system diffuse large B cell lymphoma (PCNS-DLBCL) is a rare and aggressive entity of diffuse large B cell lymphoma (DLBCL). Elements of the tumour microenvironment (TME) including tumour-infiltrating lymphocytes (TILs) and tumour-associated macrophages (TAMs) have been associated with survival in DLBCL but their composition and prognostic impact in PCNS-DLBCL are unknown. Programmed cell death-1 (PD1)/programmed death-ligand 1 (PD-L1) immune checkpoint may represent a therapeutic option. Here, we aimed to characterise PD1/PDL1 immune checkpoints and the composition of the TME in PCNS-DLBCL. We collected tumour tissue and clinical data from 57 PCNS-DLBCL and used immunohistochemistry to examine TAMs (CD68, CD163), TILs (CD3, CD4, CD8, PD1) and tumour B cells (PAX5/PDL1 double stains, PDL1). The PDL1 gene was evaluated by fluorescence in situ hybridization (FISH). PAX5/PDL1 identified PDL1 expression by tumour B cells in 10/57 cases (17.5%). PDL1 gene translocation was a recurrent cytogenetic alteration in PNCS-DLBCL (8/47.17%) and was correlated with PDL1 positive expression in tumour B cells. The TME consisted predominantly of CD163 (+) M2 TAMs and CD8 (+) TILs. Most TAMs expressed PDL1 and most TILs expressed PD1. The density of TAMs and TILs did not associate with outcome. We showed that expression of PD1 on TILs and PDL1 on TAMs, but not the expression of PDL1 on tumour B cells was correlated with better prognosis. These findings support a significant role of TME composition and PD1/PDL1 crosstalk in PCNS-DLBCL pathogenesis and bring new insights to the targeted therapy of this aggressive lymphoma.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Central Nervous System Neoplasms/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Female , France , Humans , In Situ Hybridization, Fluorescence , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma, Large B-Cell, Diffuse/diagnosis , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Macrophages/pathology , Male , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/genetics , Retrospective Studies
6.
Virchows Arch ; 475(2): 163-174, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30953147

ABSTRACT

Despite distinct clinical presentation and outcome, systemic, primary cutaneous, and breast implant-associated anaplastic large cell lymphomas (S-, PC-, BI-ALCL) ALK-negative (ALK-) show similar histopathological features including the presence of the "hallmark" cells with horseshoe-shaped nuclei and CD30 protein expression. The purpose was to better characterize these three entities using immunohistochemistry and FISH (Fluorescent in situ hybridization) to identify biomarkers differently expressed and that might be involved in their pathogenesis. Twenty-two S-ALCL ALK-, 13 PC-ALCL, and 2 BI-ALCL were included. Cases were tested for P53, P63, MUM1, MYC, GATA3, p-STAT3, PD1, and PDL1 protein expression and DUP22, TP53, TP63, MYC, and PDL1 chromosomal aberrations. As expected, S-ALCL ALK- patients had adverse outcome compare to PC and BI-ALCL. No difference was observed between the three groups concerning protein expression except for MUM1 that was significantly more frequently expressed in S-ALCL ALK- compared to PC-ALCL. In particular, constitutive activation of the STAT3 pathway and PDL1/PD1 immune-checkpoint expression was present in the three entities. TP53 deletion and PDL1 gene amplification were the commonest cytogenetic alterations and were present in the three entities. None of the studied biological parameters was associated with prognosis. Despite distinct clinical behavior, S-ALCL ALK-, PC-ALCL, and BI-ALCL share similar biological features. Larger series should be investigated with the current approach to determine more precisely the activity and the prognostic value of these biomarkers and pathways in each group.


Subject(s)
Biomarkers, Tumor/analysis , Lymphoma, Large-Cell, Anaplastic/etiology , Lymphoma, Large-Cell, Anaplastic/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Breast Implants/adverse effects , Child , Female , Humans , Lymphoma, Large-Cell, Anaplastic/metabolism , Male , Middle Aged , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Young Adult
7.
J Cutan Pathol ; 46(2): 102-110, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30370547

ABSTRACT

BACKGROUND: We report the cases of three patients presenting skin lesions whose biopsies showed nodular polymorphic infiltrates consisting of lymphocytes, plasma cells, histiocytes, eosinophils, B blasts, and Hodgkin Reed-Sternberg (HRS)-like cells. Two of them were initially diagnosed as classical Hodgkin lymphoma (cHL), on the other hand, the last one as a B-cell lymphoma. All patients have been treated for angioimmunoblastic T-cell lymphoma (AITL). METHODS: We performed a second review of the skin biopsies with further immunophenotypic molecular analyses. Scrupulous observation revealed, in the background of the three cases, atypical small to medium-sized lymphocytes carrying a CD3+, CD4+ T-cell phenotype and expressing PD1 and CXCL13 follicular helper T-cell markers. The two lesions initially diagnosed as cHL showed scattered HRS-like cells with CD30+, CD15+, PAX5+, CD20-, Epstein Barr Virus (EBV) + classical phenotype. The case initially diagnosed as B-cell lymphoma showed a diffuse B-cell proliferation associated with small B-cell and medium to large-sized B blasts that were positive for EBV. CONCLUSION: Those cases highlighted that atypical T-cells may be obscured by B-cell proliferation mimicking cHL or B-cell lymphoma in cutaneous localization of AITL and confirmed the requirement of collecting clinical information before performing a diagnosis.


Subject(s)
Hodgkin Disease , Lymphoma, B-Cell , Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Adult , Aged , Biopsy , Diagnosis, Differential , Hodgkin Disease/diagnosis , Hodgkin Disease/metabolism , Hodgkin Disease/pathology , Humans , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, T-Cell, Cutaneous/diagnosis , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Male , Middle Aged , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
8.
J Cutan Pathol ; 45(8): 610-614, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29660158

ABSTRACT

Acute myeloid leukemia (AML) may initially present as cutaneous lesions corresponding to blasts involving the skin as the first clinical manifestation prior to blood and bone marrow (BM) infiltration. Such presentation is known as myeloid leukemia cutis (LC). Blastic plasmocytoid dendritic cell neoplasm (BPDCN) is an aggressive tumor derived from the precursors of plasmocytoid dendritic cells with cutaneous and BM involvement and leukemic dissemination. Myeloid LC and BPDCN may be difficult to distinguish as they share similar clinical and histopathological features, in particular AML with monocytic differentiation. Nevertheless, the correct diagnosis has to be made to determine adequate and effective therapy. Here, we report the case of a 61-year-old woman who presented with an AML with MLL rearrangement and CD4+/CD56+ expression presenting as LC and that was misdiagnosed as BPDCN. We emphasize that careful and exhaustive analyses should be performed to make the correct diagnosis.


Subject(s)
CD4 Antigens/metabolism , CD56 Antigen/metabolism , Leukemia, Myeloid, Acute/diagnosis , Skin Neoplasms/diagnosis , Skin/pathology , Biomarkers, Tumor/metabolism , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diagnostic Errors , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Middle Aged , Skin/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
9.
Histopathology ; 71(4): 648-660, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28594133

ABSTRACT

AIMS: Distinction between primary cutaneous follicular lymphoma (PCFL) and primary cutaneous marginal zone lymphoma (PCMZL) is challenging, as clear-cut immunophenotypical and cytogenetic criteria to segregate both entities are lacking. METHODS AND RESULTS: To characterize PCFL and PCMZL more clearly and to define criteria helpful for the differential diagnosis, we compared expression of immunohistochemical markers [LIM-only transcription factor 2 (LMO2), human germinal centre-associated lymphoma (HGAL), stathmin 1 (STMN1), activation-induced cytidine deaminase (AID), myeloid cell nuclear differentiation antigen (MNDA)] and the presence of cytogenetic abnormalities described previously in nodal follicular lymphoma [B cell lymphoma 2 (BCL2) and BCL6 breaks, 1p36 chromosomal region deletion (del 1p36)] in a series of 48 cutaneous follicular and marginal zone lymphomas [cutaneous follicular lymphoma (CFL) and cutaneous marginal zone lymphoma (CMZL)]. Immunostaining for STMN1, LMO2, HGAL and AID allowed the distinction between CFL and CMZL, and STMN1 was the most sensitive marker (100% CFL, 0% CMZL). LMO2, HGAL and AID were positive in 93.2%, 82.1% and 86.2% CFL (all CMZL-negative). MNDA was expressed in both entities without significant difference (10.3% CFL, 30.8% CMZL, P = 0.18). BCL2, BCL6 breaks and the del 1p36 were present in 16.7%, 10.7% and 18.5% CFL and no CMZL. Finally, three and 29 CFL were reclassified as secondary cutaneous follicular lymphomas (SCFL) and PCFL without significant differences concerning phenotypical and cytogenetic features. BCL2, BCL6 breaks and the del 1p36 were present in 11.1%, 8% and 16.7% PCFL and did not impact the prognosis. CONCLUSION: LMO2, HGAL, STMN1 and AID, but not MNDA, are discriminant for the recognition between CFL and CMZL. BCL2, BCL6 rearrangements and the del 1p36 have a role in the pathogenesis of PCFL, the latest being the most common alteration.


Subject(s)
Biomarkers, Tumor/genetics , Chromosomes, Human, Pair 1/genetics , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, B-Cell/genetics , Lymphoma, Follicular/genetics , Skin Neoplasms/genetics , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Aged, 80 and over , Chromosome Deletion , Cytidine Deaminase/genetics , Female , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins/genetics , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell, Marginal Zone/pathology , Lymphoma, Follicular/pathology , Male , Microfilament Proteins , Middle Aged , Neoplasm Proteins/genetics , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Skin Neoplasms/pathology , Stathmin/genetics
10.
Hum Mol Genet ; 26(2): 367-382, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28365779

ABSTRACT

The cellular and molecular mechanisms underlying neurodevelopmental conditions such as autism spectrum disorders have been studied intensively for decades. The ability to generate patient-specific induced pluripotent stem cells (iPSCs) now offers a novel strategy for modelling human diseases. Recent studies have reported the derivation of iPSCs from patients with neurological disorders. The key challenge remains the demonstration of disease-related phenotypes and the ability to model the disease. Here we report a case study with signs of neurodevelopmental disorders (NDDs) harbouring chromosomal rearrangements that were sequenced using long-insert DNA paired-end tag (DNA-PET) sequencing approach. We identified the disruption of a specific gene, GTDC1. By deriving iPSCs from this patient and differentiating them into neural progenitor cells (NPCs) and neurons we dissected the disease process at the cellular level and observed defects in both NPCs and neuronal cells. We also showed that disruption of GTDC1 expression in wild type human NPCs and neurons showed a similar phenotype as patient's iPSCs. Finally, we utilized a zebrafish model to demonstrate a role for GTDC1 in the development of the central nervous system. Our findings highlight the importance of combining sequencing technologies with the iPSC technology for NDDs modelling that could be applied for personalized medicine.


Subject(s)
Autism Spectrum Disorder/genetics , Glycosyltransferases/genetics , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Animals , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , Cell Differentiation/genetics , Central Nervous System/growth & development , Central Nervous System/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Genome, Human , Glycosyltransferases/biosynthesis , High-Throughput Nucleotide Sequencing , Humans , Induced Pluripotent Stem Cells/pathology , Neural Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , Precision Medicine , Zebrafish/genetics , Zebrafish/growth & development
11.
Hematol Oncol ; 35(4): 487-496, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27966264

ABSTRACT

Primary central nervous system diffuse large B-cell lymphoma (PCNS-DLBCL) is a rare and aggressive type of diffuse large B-cell lymphoma (DLBCL) whit poorly understood pathogenesis. Finding biomarkers associated with patient survival may be important for understanding its physiopathology and to develop new therapeutic approaches. We investigated 32 PCNS-DLBCL from immunocompetent patients for BCL2, CMYC, LMO2, and P53 expression and for cytogenetic aberrations of BCL2, BCL6, and MYC genes, all known for their prognostic value in systemic DLBCL (s-DLBCL). We analyzed PD1 and PDL1 protein expression in both tumor infiltrating lymphocytes (TILs) and tumor cells. Finally, we searched for correlation between biological data and clinical course. The PCNS-DLBCL expressed BCL2, CMYC, LMO2, and P53 at similar frequency than s-DLBCL but without significant prognostic on survival. None cases harbored aberrations involving BCL2 and MYC gene whereas BCL6 abnormalities were present in 20.7% of cases but without value on survival. Expression of PD1 in TILs and PDL1 in tumor cells was observed at higher rates than in s-DLBCL (58% and 37%, respectively). The PD1 expression in TILs correlated with PDL1 expression in tumor cells (P = .001). Presence of PD1 positive TILs was associated with poorer overall survival (P = .011). Patients with PDL1 overexpression tended to better response to chemotherapy (P = .23). In conclusion PCNS-DLBCL pathogenesis differs from s-DLBCL without prognostic value of the phenotypic and cytogenetic parameters known for their pejorative impact in the latter. The PD1/PDL1 pathway plays a strong role in PCNS-DLBCL and represents an attractive target for this aggressive lymphoma.


Subject(s)
B7-H1 Antigen/metabolism , Central Nervous System Neoplasms/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Programmed Cell Death 1 Receptor/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Central Nervous System Neoplasms/mortality , Central Nervous System Neoplasms/pathology , Female , Humans , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Retrospective Studies
12.
Virchows Arch ; 469(6): 697-706, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27605053

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.


Subject(s)
Chromosomes, Human, Pair 13 , Lymphoma, Mantle-Cell/diagnosis , Lymphoma, Mantle-Cell/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Translocation, Genetic/genetics , Aged , Aged, 80 and over , Chromosome Deletion , Comparative Genomic Hybridization/methods , Female , Humans , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Lymphoma, Mantle-Cell/pathology , Male , Middle Aged , Phenotype , Prognosis
13.
Eur J Med Genet ; 59(3): 173-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26855057

ABSTRACT

Noonan syndrome is associated with a range of malignancies including acute lymphoblastic leukemia (ALL). However, little information is available regarding the frequency, natural history, characteristics and prognosis of ALL in Noonan syndrome or RASopathies in general. Cross-referencing data from a large prospective cohort of 1176 patients having a molecularly confirmed RASopathy with data from the French childhood cancer registry allowed us to identify ALL in 6 (0.5%) patients including 4/778 (0.5%) with a germline PTPN11 mutation and 2/94 (2.1%) with a germline SOS1 mutation. None of the patients of our series with CFC syndrome (with germline BRAF or MAP2K1/MAP2K2 mutation - n = 121) or Costello syndrome (with HRAS mutation - n = 35) had an ALL. A total of 19 Noonan-ALL were gathered by adding our patients to those of the International Berlin-Munster-Frankfurt (I-BFM) study group and previously reported patients. Strikingly, all Noonan-associated ALL were B-cell precursor ALL, and high hyperdiploidy with more than 50 chromosomes was found in the leukemia cells of 13/17 (76%) patients with available genetics data. Our data suggest that children with Noonan syndrome are at higher risk to develop ALL. Like what is observed for somatic PTPN11 mutations, NS is preferentially associated with the development of hyperdiploid ALL that will usually respond well to chemotherapy. However, Noonan syndrome patients seem to have a propensity to develop post therapy myelodysplasia that can eventually be fatal. Hence, one should be particularly cautious when treating these patients.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , ras Proteins/genetics , Adolescent , Child , Child, Preschool , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Mutation , Neoplasms, Second Primary/etiology , Noonan Syndrome/complications , Noonan Syndrome/genetics , Noonan Syndrome/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Prevalence , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , SOS1 Protein/genetics , ras Proteins/metabolism
14.
Hum Mutat ; 35(11): 1311-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25137640

ABSTRACT

MED13L is a component subunit of the Mediator complex, an important regulator of transcription that is highly conserved across eukaryotes. Here, we report MED13L disruption in a translocation t(12;19) breakpoint of a patient with Pierre-Robin syndrome, moderate intellectual disability, craniofacial anomalies, and muscular defects. The phenotype is similar to previously described patients with MED13L haploinsufficiency. Knockdown of MED13L orthologue in zebrafish, med13b, showed early defective migration of cranial neural crest cells (NCCs) that contributed to cartilage structure deformities in the later stage, recapitulating craniofacial anomalies seen in human patients. Notably, we observed abnormal distribution of developing neurons in different brain regions of med13b morphant embryos, which could be rescued upon introduction of full-length human MED13L mRNA. To compare with mammalian system, we suppressed MED13L expression by short-hairpin RNA in ES-derived human neural progenitors, and differentiated them into neurons. Transcriptome analysis revealed differential expression of components of Wnt and FGF signaling pathways in MED13L-deficient neurons. Our finding provides a novel insight into the mechanism of overlapping phenotypic outcome targeting NCCs derivatives organs in patients with MED13L haploinsufficiency, and emphasizes a clinically recognizable syndromic phenotype in these patients.


Subject(s)
Haploinsufficiency , Intellectual Disability/genetics , Mediator Complex/genetics , Neural Crest/metabolism , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Child, Preschool , Chromosome Breakpoints , Disease Models, Animal , Embryonic Stem Cells/metabolism , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genetic Association Studies , Humans , Intellectual Disability/diagnosis , Mediator Complex/metabolism , Neural Crest/embryology , Neurons/cytology , Neurons/metabolism , Phenotype , RNA, Messenger/genetics , Sequence Analysis, DNA , Transcriptome , Translocation, Genetic , Zebrafish
15.
PLoS One ; 9(6): e90852, 2014.
Article in English | MEDLINE | ID: mdl-24603971

ABSTRACT

Delineating candidate genes at the chromosomal breakpoint regions in the apparently balanced chromosome rearrangements (ABCR) has been shown to be more effective with the emergence of next-generation sequencing (NGS) technologies. We employed a large-insert (7-11 kb) paired-end tag sequencing technology (DNA-PET) to systematically analyze genome of four patients harbouring cytogenetically defined ABCR with neurodevelopmental symptoms, including developmental delay (DD) and speech disorders. We characterized structural variants (SVs) specific to each individual, including those matching the chromosomal breakpoints. Refinement of these regions by Sanger sequencing resulted in the identification of five disrupted genes in three individuals: guanine nucleotide binding protein, q polypeptide (GNAQ), RNA-binding protein, fox-1 homolog (RBFOX3), unc-5 homolog D (C.elegans) (UNC5D), transmembrane protein 47 (TMEM47), and X-linked inhibitor of apoptosis (XIAP). Among them, XIAP is the causative gene for the immunodeficiency phenotype seen in the patient. The remaining genes displayed specific expression in the fetal brain and have known biologically relevant functions in brain development, suggesting putative candidate genes for neurodevelopmental phenotypes. This study demonstrates the application of NGS technologies in mapping individual gene disruptions in ABCR as a resource for deciphering candidate genes in human neurodevelopmental disorders (NDDs).


Subject(s)
Chromosome Breakpoints , Developmental Disabilities/genetics , Language Development Disorders/genetics , Base Sequence , Chromosome Inversion , DNA Copy Number Variations , Female , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Male , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA , Translocation, Genetic
16.
Eur J Med Genet ; 56(11): 635-41, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24013099

ABSTRACT

7qter deletion syndrome includes prenatal and/or postnatal growth retardation, microcephaly, psychomotor delay or mental retardation and a characteristic dysmorphism. If clinical features are well described, the molecular mechanisms underlying the 7qter deletion syndrome remain unknown. Those deletions usually arise de novo. Here, we describe a young boy with an abnormal phenotype consistent with a 7qter deletion syndrome. High resolution genomic analysis (Affymetrix Human Genome Wide SNP 6.0) revealed a 7q36.3 deletion encompassing NCAPG2, ESYT2, WDR60 and VIPR2, inherited from his asymptomatic father and paternal grandfather. In addition, the patient also harbored a MCPH1 deletion inherited from his healthy mother. Combined NCAPG2 and MCPH1 deletions were correlated with low mRNA levels and protein expression in the patient. MCPH1 and NCAPG2 proteins interaction is known to control chromosome structure and we thus propose that double heterozygosity for null mutations of those two genes of the Condensin II system contribute to mental deficiency with severe microcephaly phenotype.


Subject(s)
Adenosine Triphosphatases/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/genetics , Gene Deletion , Intellectual Disability/genetics , Microcephaly/genetics , Multiprotein Complexes/genetics , Nerve Tissue Proteins/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins , Child , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human, Pair 7/genetics , Cytoskeletal Proteins , DNA-Binding Proteins/metabolism , Genetic Loci/genetics , Heterozygote , Humans , Intellectual Disability/diagnosis , Male , Microcephaly/diagnosis , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Pedigree , Syndrome
17.
Sci Transl Med ; 3(77): 77ra30, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21471434

ABSTRACT

Fusion genes are chimeric genes formed in cancers through genomic aberrations such as translocations, amplifications, and rearrangements. To identify fusion genes in gastric cancer, we analyzed regions of chromosomal imbalance in a cohort of 106 primary gastric cancers and 27 cell lines derived from gastric cancers. Multiple samples exhibited genomic breakpoints in the 5' region of SLC1A2/EAAT2, a gene encoding a glutamate transporter. Analysis of a breakpoint-positive SNU16 cell line revealed expression of a CD44-SLC1A2 fusion transcript caused by a paracentric chromosomal inversion, which was predicted to produce a truncated but functional SLC1A2 protein. In primary tumors, CD44-SLC1A2 gene fusions were detected in 1 to 2% of gastric cancers, but not in adjacent matched normal gastric tissues. When we specifically silenced CD44-SLC1A2, cellular proliferation, invasion, and anchorage-independent growth were significantly reduced. Conversely, CD44-SLC1A2 overexpression in gastric cells stimulated these pro-oncogenic traits. CD44-SLC1A2 silencing caused significant reductions in intracellular glutamate concentrations and sensitized SNU16 cells to cisplatin, a commonly used chemotherapeutic agent in gastric cancer. We conclude that fusion of the SLC1A2 gene coding region to CD44 regulatory elements likely causes SLC1A2 transcriptional dysregulation, because tumors expressing high SLC1A2 levels also tended to be CD44-SLC1A2-positive. CD44-SLC1A2 may represent a class of gene fusions in cancers that establish a pro-oncogenic metabolic milieu favoring tumor growth and survival.


Subject(s)
Gene Fusion/genetics , Glutamate Plasma Membrane Transport Proteins/genetics , Hyaluronan Receptors/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Excitatory Amino Acid Transporter 2 , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Arch Pathol Lab Med ; 132(6): 965-73, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18517280

ABSTRACT

CONTEXT: Deletion of the short arm of chromosome 3 (3p deletion) is a cytogenetic abnormality generally associated with clear cell renal cell carcinoma, the most aggressive form of renal epithelial tumor. OBJECTIVE: To cytogenetically characterize 24 renal tumors in order to check the incidence and the type of 3p deletions, as well as to identify new genes putatively participating in renal tumorigenesis and test the protein products of the von Hippel-Lindau (VHL) and fragile histidine triad (FHIT) genes. DESIGN: We analyzed 24 renal tumors by conventional cytogenetics, comparative genomic hybridization, and fluorescence in situ hybridization. We then performed a comparative expression study of the proteins pVHL and Fhit. RESULTS: In our series of 24 renal tumors, the 3p deletion was the most frequent genetic alteration (15/24); the other features were partial trisomy 5q, 8p deletion, and monosomy 9 and 14. The 3p deletion was long and terminal, and no interstitial deletion was identified. By immunohistochemistry, we found that for the 2 genes of interest, VHL and FHIT, loss or decrease in protein expression were very frequently observed in clear cell renal cell carcinoma and not always associated with a 3p deletion (14/20 in clear cell renal cell carcinoma). CONCLUSIONS: Our studies characterize the 3p deletion as long and terminal and identify no interstitial deletion in that chromosome. Fhit and pVHL protein expression loss appear to be independent, as they can be dissociated. Our data are supportive of a role for FHIT (in addition to VHL) in renal tumorigenesis. No other gene with particular potential interest in renal tumorigenesis could be identified among the selected genes.


Subject(s)
Acid Anhydride Hydrolases/genetics , Carcinoma, Renal Cell/genetics , Chromosomes, Human, Pair 3/genetics , Kidney Neoplasms/genetics , Neoplasm Proteins/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Chromosome Aberrations , Chromosome Deletion , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged
19.
Neurobiol Dis ; 15(2): 240-50, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006694

ABSTRACT

ZFHX1B encodes Smad-interacting protein 1, a transcriptional corepressor involved in the transforming growth factors beta (TGFbeta) signaling pathway. ZFHX1B mutations cause a complex developmental phenotype characterized by severe mental retardation (MR) and multiple congenital defects. We compared the distribution of ZFHX1B transcripts during mouse and human embryogenesis as well as in adult mice and humans. This showed that this gene is strongly transcribed at an early stage in the developing peripheral and central nervous systems of both mice and humans, in all neuronal regions of the brains of 25-week human fetuses and adult mice, and at varying levels in numerous nonneural tissues. Northern blot analysis suggested that ZFHX1B undergoes tissue-specific alternative splicing in both species. These results strongly suggest that ZFHX1B determines the transcriptional levels of target genes in various tissues through the combinatorial interactions of its isoforms with different Smad proteins. Thus, as well as causing neural defects, ZFHX1B mutations may also cause other malformations.


Subject(s)
Brain/abnormalities , Brain/metabolism , Embryo, Mammalian/abnormalities , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Intellectual Disability/genetics , Repressor Proteins/metabolism , Alternative Splicing/genetics , Animals , Body Patterning/genetics , Brain/physiopathology , Congenital Abnormalities/genetics , Congenital Abnormalities/metabolism , Congenital Abnormalities/physiopathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/physiopathology , DNA-Binding Proteins/metabolism , Embryo, Mammalian/physiopathology , Fetus , Genes, Regulator/genetics , Homeodomain Proteins/genetics , Humans , Intellectual Disability/metabolism , Intellectual Disability/physiopathology , Mice , Mutation/genetics , Neural Crest/abnormalities , Neural Crest/metabolism , Neural Crest/physiopathology , Protein Isoforms/genetics , Repressor Proteins/genetics , Smad Proteins , Trans-Activators/metabolism , Zinc Finger E-box Binding Homeobox 2
20.
Am J Med Genet A ; 119A(3): 257-65, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12784289

ABSTRACT

Mutations or deletions involving ZFHX1B (previously SIP1) have recently been found to cause one form of syndromic Hirschsprung disease (HSCR), associated with microcephaly, mental retardation, and distinctive facial features. Patients with the characteristic facial phenotype and severe mental retardation, but without HSCR, have now also been shown to have mutations in this gene. Mutations of ZFHX1B are frequently associated with other congenital anomalies, including congenital heart disease, hypospadias, renal tract anomalies, and agenesis of the corpus callosum (ACC). We present the clinical data and mutation analysis results from a series of 23 patients with this clinical syndrome, of whom 21 have proven ZFHX1B mutations or deletions (15 previously unpublished). Two patients with the typical features (one with and one without HSCR) did not have detectable abnormalities of ZFHX1B. We emphasize that this syndrome can be recognized by the facial phenotype in the absence of either HSCR or other congenital anomalies, and needs to be considered in the differential diagnosis of dysmorphism with severe mental retardation +/- epilepsy.


Subject(s)
Abnormalities, Multiple/genetics , Face/abnormalities , Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Intellectual Disability/genetics , Mutation/genetics , Repressor Proteins/genetics , Child , Child, Preschool , DNA Mutational Analysis , DNA Probes , Epilepsy/genetics , Female , Heart Diseases/congenital , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Microcephaly/genetics , Phenotype , Polymerase Chain Reaction , Zinc Finger E-box Binding Homeobox 2 , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...