Genetic alterations and oxidative stress in T cell lymphomas

2022 ◽  
pp. 108109
Author(s):  
Sushant Kumar ◽  
Bhavuk Dhamija ◽  
Diksha Attrish ◽  
Vinanti Sawant ◽  
Manju Sengar ◽  
...  
2020 ◽  
Vol 4 (10) ◽  
pp. 2261-2271 ◽  
Author(s):  
Natasha E. Lewis ◽  
Kseniya Petrova-Drus ◽  
Sarah Huet ◽  
Zachary D. Epstein-Peterson ◽  
Qi Gao ◽  
...  

Abstract TET2 and DNMT3A mutations are frequently identified in T-cell lymphomas of T follicular helper cell origin (TCL-TFH), clonal hematopoiesis (CH), and myeloid neoplasms (MNs). The relationships among these 3 entities, however, are not well understood. We performed comprehensive genomic studies on paired bone marrow and tissue samples as well as on flow cytometry–sorted bone marrow and peripheral blood subpopulations from a cohort of 22 patients with TCL-TFH to identify shared CH-type mutations in various hematopoietic cell compartments. Identical mutations were detected in the neoplastic T-cell and myeloid compartments of 15 out of 22 patients (68%), including TET2 (14/15) and DNMT3A (10/15). Four patients developed MNs, all of which shared CH-type mutations with their TCL-TFH; additional unique genetic alterations were also detected in each patient’s TCL-TFH and MN. These data demonstrate that CH is prevalent in patients with TCL-TFH and that divergent evolution of a CH clone may give rise to both TCL-TFH and MNs.


Author(s):  
Jennifer Mier-Cabrera ◽  
Luis Jiménez-Zamudio ◽  
Ethel García-Latorre ◽  
Oliver Cruz-Orozco ◽  
César Hernández-Guerrero

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4211-4211
Author(s):  
Shaker A. Mousa ◽  
Ghanshyam Patil ◽  
Abdelhadi Rebbaa

Abstract The development of resistance to chemotherapy represents an adaptive biological response by tumor cells that leads to treatment failure and patient relapse. During the course of their evolution (intrinsic resistance) or in response to chemotherapy (acquired resistance), tumor cells may undergo genetic alterations to possess a drug resistant phenotype. Dysregulation of membrane transport proteins and cellular enzymes, as well as altered susceptibility to commit to apoptosis are among the mechanisms that contribute to the genesis of acquired drug resistance. Recently, the development of approaches to prevent and/or to reverse this phenomenon has attracted special interest and a number of drug candidates have been identified. Despite strong effects observed for these candidates in vitro, however, most of them fail in vivo. In the present study, we have identified a novel small molecule inhibitor of dual NF-κB and oxidative stress pathways, OT-304, as a potential candidate to reverse drug resistance. Initial investigations indicate that this compound effectively inhibits proliferation of doxorubicin-sensitive and doxorubicin-resistant cells to the same extent, suggesting that it is capable of bypassing the development of drug resistance. Additional experiments reveal that OT-304 enhances cancer cell sensitivity to doxorubicin and to etoposide, particularly in cells characterized by the over-expression of the drug transporter P-glycoprotein. These findings suggest that either the expression/and or the function of P-glycoprotein could be affected by OT-304. In vivo studies using tumor xenografts in nude mice showed that OT-304 is also capable of preventing the growth of drug resistant cancer cells. This later finding further confirms the role of OT-304 as a drug resistance-reversing agent and warrants further pre-clinical and clinical investigation to determine its efficacy in treating aggressive tumors.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2927-2927 ◽  
Author(s):  
Yosaku Watatani ◽  
Yasuharu Sato ◽  
Kenji Nishida ◽  
Hiroaki Miyoshi ◽  
Yuichi Shiraishi ◽  
...  

Abstract Peripheral T-cell lymphomas (PTCLs) are a heterogeneous group of lymphoproliferative disorders arising from mature T-cells. Among them, PTCL-not otherwise specified (PTCL-NOS) is a diagnosis of exclusion, comprising the largest fraction of PTCL with a diverse underlying pathogenesis. Recently, the concept of nodal T-cell lymphomas with T-follicular helper (TFH) phenotype, including angioimmunoblastic T-cell lymphoma (AITL) and PTCL-NOS that manifests a TFH phenotype, has been proposed, a distinguishing feature of which is the high frequency of TET2, IDH2, DNMT3A, and RHOA(G17V) mutations. Although recent large-scale genetic studies have uncovered mutational landscapes of several other subtypes of PTCLs, such as cutaneous T-cell lymphoma and adult T-cell leukemia/lymphoma (ATL), the entire picture of somatic alterations in PTCL-NOS still remains elusive. In addition, their similarities and differences among various histological subtypes in PTCLs have not been fully elucidated. To address this issue, we initially analyzed our and publicly available whole-exome/genome as well as transcriptome sequencing data from PTCL-NOS and other related PTCLs. Then, we carried out an extensive investigation of somatic mutations and structural variations (SVs) in PTCL-NOS using targeted-capture sequencing of 118 PTCL-NOS samples. Consistent with previous reports, TET2 (35%) was the most frequently mutated gene in PTCL-NOS with the majority (78%) affected by multiple mutations, followed by RHOA (25%), TP53 (16%), KMT2C (12%), PLCG1 (12%), and HLA-B (11%). Besides them, a considerable proportion of patients harbored mutations in components of T-cell receptor (TCR) /NF-κB pathway (such as PRKCB, CARD11, IRF4, and PRDM1), other signal transduction molecules (STAT3, NOTCH1, and SOCS1), chemokine receptors (CCR4 and CCR7), epigenetic modifiers (CREBBP, KDM6A, IDH2, and DNMT3A), transcriptional regulators (GATA3 and TBL1XR1), and molecules associated with immune evasion (HLA-A, HLA-B, FAS, B2M, and CD58). In addition to deteriorating SVs involving frequently affected genes (TP53, FAS, GATA3, and TBL1XR1), we discovered several genes almost exclusively affected by SVs, including TP73, IKZF2, and NFKB2, and CD274. Novel targets of recurrent mutation were also identified, including PDCD1, YTHDF2, and LRP1B, which were frequently targeted by nonsense and frameshift mutations distributed throughout the entire genes. Among them, PDCD1encodes PD-1 receptor transmitting an inhibitory signal from PD-L1 and PD-L2 ligands in T cells, and its loss of function seems to enable tumor cells to escape from the suppression by this negative signal. Although the roles of YTHDF2, a reader protein of N6-methyladenosine, and LRP1B, a member of the low density lipoprotein receptor family, in T cells are not immediately apparent, these findings shed light on a new biological function of these genes. Next, we investigated the co-existence relationship between frequently altered genes in PTCL-NOS. Interestingly, mutations characteristic of TFH lymphomas (TET2, RHOA, IDH2, and DNMT3A) tended to co-occur in a subset of PTCL-NOS cases, whereas they were almost mutually exclusive with mutations in TP53 and TCR/NF-κB pathway genes. This observation reveals the molecular distinction between TFH and non-TFH lymphomas in PTCL-NOS: the former is similar to AITL, although TET2 mutations did not show higher allelic burden than RHOA and IDH2mutations. In contrast, the latter is at least partly characterized by the genetic alterations shared with ATL. In summary, our findings illuminate the landscape of somatic alterations in PTCL-NOS and provide a novel insight into their genetic and molecular heterogeneity, which would help us to exploit a new therapeutic strategy to combat this disease. Disclosures Ohshima: CHUGAI PHARMACEUTICAL CO.,LTD.: Research Funding, Speakers Bureau; Kyowa Hakko Kirin Co., Ltd.: Research Funding, Speakers Bureau. Ogawa:Kan research institute: Consultancy, Research Funding; Sumitomo Dainippon Pharma: Research Funding; Takeda Pharmaceuticals: Consultancy, Research Funding. Kataoka:Kyowa Hakko Kirin: Honoraria; Yakult: Honoraria; Boehringer Ingelheim: Honoraria.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1482-1482
Author(s):  
Jose Rodriguez Cortes ◽  
Anisha R. Cooke ◽  
Aidan Quinn ◽  
Ioan Filip ◽  
Juan Angel Patino ◽  
...  

Peripheral T-cell lymphomas (PTCL) are highly aggressive, malignant hematologic tumors that arise from clonal proliferation of mature post-thymic T-cells. Among these, angioimmunoblastic T-cell lymphoma (AITL), and peripheral T-cell lymphomas not otherwise specified (PTCL, NOS) account for >45% diagnoses, show limited response to intensified chemotherapy treatment with dismal overall survival rate. Our genomic studies have uncovered recurrent mutations and novel cancer-associated gene fusions involving the guanine nucleotide exchange factor VAV1 in AITL and PTCL, NOS, supporting a role for the VAV1 oncogene in the pathogenesis of PTCL. Genetic alterations in VAV1 found in PTCL included recurrent deletions resulting in the in-frame deletion of 9 codons (DV778_T786) in the linker region connecting the C-terminal SH2 domain to the downstream SH3 domain on VAV1 as well as recurrent different gene-fusion transcripts encoding proteins in which the C-terminal SH3 domain of VAV1 is replaced by different domains of the fusion partner. Consistent with the auto-inhibitory role of the C-SH3 domain, the PTCL-associated VAV1 alterations resulted in increased VAV1 activity and strong activation of the mitogen-activated protein kinase (ERK1/2), c-Jun N-terminal kinase (JNK) and nuclear factor of activated T-cells (NFAT) pathways. Interestingly, mutation co-occurrence analysis shows significant mutual exclusivity of the highly prevalent AITL RHOA G17V mutation and VAV1 genomic alterations (p-value 0.0142) in AITL and PTCL, NOS. This observation supports that VAV1 genetic alterations and RHOA G17V could share a common mechanism of action. To study the role and mechanisms of VAV1 alterations in PTCL in vivo we have engineered a C57/BL6 mouse lines with conditional expression of the recurrent Vav1-Myo1f fusion. By crossing Vav1-myo1f conditional knockin with CD4-CreERT2 mouse deletor line, we have generated animals with CD4+ T cell-specific and tamoxifen-inducible expression of Vav1-myo1f. Preliminary studies indicate that expression of Vav1-myo1f in CD4+ cells induces expansion of TFH cells. This phenotype partially resembles that of animals with CD4 specific expression of Rhoa G17V, indicating a potential convergent effect and mechanisms for VAV1 alterations and RHOA G17V in T-cell development and transformation. However, and more interestingly, Vav1-myo1f expressing cells specifically present a memory cell-associated immunophenotype (CD44+ CD62L-) and characteristic Th2-like features that had not been observed in the Rhoa G17V model. Analysis of signaling pathways activated in control and Vav1-myo1f expressing CD4+ cells showed increased phosphorylation of Vav1 and increased activation of the MAPK pathway upon expression of Vav1-myo1f, supporting the role of Vav1 alterations as regulators of T-cell activation. Notably, single cell transcriptome analysis of bulk splenic cells isolated from CD4-CreERT2 control and CD4-CreERT2Vav1co-Vav1-myo1f/WT mice indicate that expression of Vav1-myo1f in CD4+ cells leads to a significant expansion of monocytes and TFH cells and a decrease in B-cell, both naïve and memory. These results support that expression of Vav1-myo1f in CD4+ cells can deeply modify the lymphoid microenvironment. Finally, preliminary analysis of a limited number of mice with targeted expression of the Vav1-Myo1f fusion in CD4+ cells (CD4-Cre Vav1co-Vav1-Myo1f /WT) showed development of a fatal CD4+ malignant lymphoproliferation with a latency of 6-14 months. Histological examination showed disrupted splenic architecture accompanied with clonal expansion of CD4+ cells indicative of T-cell lymphoma with PTCL, NOS characteristics and verified expression of both wild type Vav1 and Vav1-myo1f alleles. Interestingly, Vav1-myo1f lymphoma cells showed Th2 associated features including expression of Gata3, c-Maf and IL4 and Il10. These results support an important role for VAV1 alterations in T-cell differentiation, remodeling of the tumor microenvironment and T-cell lymphoma development and highlight the importance of our model as a new experimental system to study the evolution of PTCL and develop novel targeted therapies for this disease. Disclosures Palomero: Kura Oncology: Research Funding.


2021 ◽  
Author(s):  
Ayumi Fujimoto ◽  
Ritsuro Suzuki

Natural killer (NK)/T cell lymphoma includes two major subtypes of disease, specifically extranodal NK/T cell lymphoma, nasal type (ENKL) and aggressive NK cell leukemia (ANKL). Both are strongly associated with Epstein-Barr virus and are prevalent in East Asia and Latin America. Except for that of limited-stage ENKL, the prognosis of both diseases was poor in the previous decade. The advent of non-anthracycline-based chemoradiotherapy has contributed to an improvement in ENKL prognosis, but there is still room for further treatment progress. Recently, the high efficacy of PD-1 antibody was reported in relapsed or refractory ENKL patients. This was later supported by the finding that PD-L1/PD-L2 genetic alterations are frequently observed in ENKL and ANKL patients. Due to the rarity of the disease, a standard treatment for ANKL remains to be established. Currently, allogeneic stem cell transplantation is the only curative treatment, and this is even applicable to chemo-resistant ANKL patients. In this review, we focus on recent treatment approaches for NK/T cell lymphomas including novel agents.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3567-3567
Author(s):  
Laurence de Leval ◽  
David Rickman ◽  
Emilie Thomas ◽  
Louis Huang ◽  
Aurélien de Reynies ◽  
...  

Abstract Genetic alterations underlying angioimmunoblastic and unspecified peripheral T-cell lymphomas (AITL and PTCL-u) are largely unknown. Seventeen AITL and 16 PTCL-u previously characterized by gene expression profiling, were analyzed by CGH on DNA microarrays comprising 4434 BAC clones with a resolution of about 600 KB. In the PTCL-u group, the mean number of chromosomal aberrations per case was 302 (range, 55 to 892). Gains (n=237, 41 to 587) were more frequent than losses (n=65, 8 to 305). AITL samples had, on average, a lesser number of genomic alterations than PTCL-u cases (n=243, range, 55 to 485), comprising more gains (n=201, 42 to 541) than losses (n=42, 9 to 262). Overall, the most frequent recurrent gains, present in 50% of all samples, were observed at 1p36.1; 1p36.3 ; 1q32 ; 2q37 ; 4p16 ; 5p15.3 ; 6q12 ; 7p22 ; 7p12 ; 7p11.2 ; 7q35-36 ; 8q24.3 ; 9q34 ; 11p15 ; 11q13 ; 16p13.3 ; 16q24 ; 17q12,q21,q25 ; 19p13.3 ; 19q13.2-q13.3 ; 20q11.2-q13.3 ; 22q11.1-q11.2 ; Xp11 ; Xp21-22 ; Xq27-28. The comparison of the genomic profiles of AITL and PTCL-u identified 73 genomic alterations (at clones or zones of clones) significantly associated with either group of tumors (Fisher test, p < 0,05). Six genomic gains mapping at 5p15 and 22q11 were associated with the AITL subtype. Thirty-four gains (mapping at 6p25, 7p1, 7q3, 8q24, 11p14, 14q32, 17q, 22q) and 33 losses (mapping at 6q, 10p and 13q), were overrepresented in PTCL-u. The coordinate analysis of the transcriptomic and CGH array data identified 10 regions with genomic imbalances containing genes differentially expressed in AITL versus PTCL-u. Seven regions amplified in PTCL-u contained genes overexpressed in PTCL-u, mostly related to metabolic pathways. Conversely, loss of genomic material at 13q12 correlated with decreased expression in PTCL-u of a few genes of the AITL signature. In AITL tumors, gain at 22q11 correlated with increased transcription of the LIF gene, previosuly characterized as part of the tumor cell signature in AITL. CD30+ PTCL-u samples had on average a higher number of genomic aberrations than CD30-negative cases (n=408 versus 238). Thirty-three genomic gains and 22 losses were exclusively seen in CD30+ tumors, and regions with chromosomal imbalances at 1q, 6q, 10p contained genes differentially expressed in CD30+ and CD30− tumors. For example, reduced transcription of FYN in CD30+ PTCL-u correlated with deletion of the corresponding chromosomal region. In conclusion, all 33 nodal PTCL analyzed harbor genomic imbalances (gains>losses), of which many are common to both AITL and PTCL-u subgroups; the pattern of genomic aberrations differs between the two subgroups, with certain aberrations being overrepresented in PTCL-u, and only a few specific for AITL; coordinate appraisal of transcriptomic and genomic data highlights correlations between genomic imbalances and gene expression signatures in subgroups of tumors.


Blood ◽  
2009 ◽  
Vol 113 (1) ◽  
pp. 127-136 ◽  
Author(s):  
Remco van Doorn ◽  
Marloes S. van Kester ◽  
Remco Dijkman ◽  
Maarten H. Vermeer ◽  
Aat A. Mulder ◽  
...  

Abstract Mycosis fungoides (MF), the most common cutaneous T-cell lymphoma, is a malignancy of mature, skin-homing T cells. Sézary syndrome (Sz) is often considered to represent a leukemic phase of MF. In this study, the pattern of numerical chromosomal alterations in MF tumor samples was defined using array-based comparative genomic hybridization (CGH); simultaneously, gene expression was analyzed using microarrays. Highly recurrent chromosomal alterations in MF include gain of 7q36, 7q21-7q22 and loss of 5q13 and 9p21. The pattern characteristic of MF differs markedly from chromosomal alterations observed in Sz. Integration of data from array-based CGH and gene-expression analysis yielded several candidate genes with potential relevance in the pathogenesis of MF. We confirmed that the FASTK and SKAP1 genes, residing in loci with recurrent gain, demonstrated increased expression. The RB1 and DLEU1 tumor suppressor genes showed diminished expression associated with loss. In addition, it was found that the presence of chromosomal alterations on 9p21, 8q24, and 1q21-1q22 was associated with poor prognosis in patients with MF. This study provides novel insight into genetic alterations underlying MF. Furthermore, our analysis uncovered genomic differences between MF and Sz, which suggest that the molecular pathogenesis and therefore therapeutic requirements of these cutaneous T-cell lymphomas may be distinct.


Sign in / Sign up

Export Citation Format

Share Document