1
|
Azizan A, Farhadi E, Faezi ST, Alikhani M, Vojgani Y, Enayati S, Jamshidi A, Vodjgani M, Mahmoudi M. Dysregulated CDKN2A-MDM2-p53 axis in B cell of systemic lupus erythematosus. Int Immunopharmacol 2025; 150:114297. [PMID: 39970716 DOI: 10.1016/j.intimp.2025.114297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/14/2024] [Accepted: 02/11/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND The complicated autoimmune disease known as systemic lupus erythematosus (SLE) is characterized by abnormal immunological B cell modulation that exhibits enhanced survival. The genes Tp53, MDM2, and CDKN2A (ARF) play crucial roles in cellular regulation. In this study, we aim to elucidate the status of these genes. This might be a factor in the B cells' higher survival rate in SLE. METHODS B cells were obtained from the peripheral blood of participants and cultured, with a subset of the cells activated using anti-IgM. The expression levels of Tp53, MDM2, and CDKN2A genes were assessed both at baseline and after activation. RESULTS Tp53 and CDKN2A exhibited decreased expression in both baseline and activated B cells, with further decreases observed active and inactive SLE. Conversely, MDM2 demonstrated increased expression in baseline B cells, with a more pronounced increase observed in activated B cells of active SLE. Correlation analysis indicated several significant positive and negative associations, particularly between disease activity indicators and MDM2 expression, as well as among the studied genes. CONCLUSION The dysregulation of the CDKN2A-MDM2-p53 axis in B cells of SLE may lead to increased B cell survival due to reduced Tp53 and CDKN2A expression and elevated MDM2 levels. In addition, MDM2 gene expression levels show a significant positive correlation with disease activity indices like anti-dsDNA titer and SLEDAI score in SLE patients.
Collapse
Affiliation(s)
- Amin Azizan
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran.
| | | | - Majid Alikhani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Vojgani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Enayati
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vodjgani
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Sarkozy C, Tessoulin B, Chiron D. Unraveling MCL biology to understand resistance and identify vulnerabilities. Blood 2025; 145:696-707. [PMID: 38551811 DOI: 10.1182/blood.2023022351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/15/2024] [Indexed: 02/14/2025] Open
Abstract
ABSTRACT Mantle cell lymphoma (MCL) is a rare (5%-7%), aggressive B-cell non-Hodgkin lymphoma with well-defined hallmarks (eg, cyclin D1, SOX11), and its expansion is highly dependent on the tumor microenvironment (TME). Parallel drastic progress in the understanding of lymphomagenesis and improved treatments led to a paradigm shift in this B-cell malignancy with now prolonged disease-free survival after intensive chemotherapy and anti-CD20-based maintenance. However, this toxic strategy is not applicable in frail or older patients, and a small but significant part of the cases present a refractory disease representing unmet medical needs. Importantly, the field has recently seen the rapid emergence of targeted and immune-based strategies with effective combinations relying on biological rationales to overcome malignant plasticity and intratumor heterogeneity. In this review, we expose how unraveling the biology of MCL allows to better understand the therapeutic resistances and to identify neo-vulnerabilities in tumors, which are essential to offer efficient novel strategies for high-risk patients. We first highlight the tumor intrinsic resistance mechanisms and associated Achilles heels within various pathways, such as NF-κB, mitochondrial apoptosis, DNA repair, and epigenetic regulators. We then place the tumor in its complex ecosystem to decipher the dialog with the multiple TME components and show how the resulting protumoral signals could be disrupted with innovative therapeutic strategies. Finally, we discuss how these progresses could be integrated into a personalized approach in MCL.
Collapse
Affiliation(s)
- Clémentine Sarkozy
- Service d'Hématologie, Institut Curie, Saint Cloud, France
- Laboratoire d'Imagerie Translationnelle en Oncologie, U1288 Inserm/Institut Curie Centre de Recherche, Paris, France
| | - Benoit Tessoulin
- Service d'Hématologie, Centre Hospitalier Universitaire Nantes, Nantes, France
- reMoVE-B, Nantes Université, INSERM, Centre National de la Recherche Scientifique, Université d'Angers, CRCI2NA, Nantes, France
| | - David Chiron
- reMoVE-B, Nantes Université, INSERM, Centre National de la Recherche Scientifique, Université d'Angers, CRCI2NA, Nantes, France
| |
Collapse
|
3
|
Durand R, Bellanger C, Descamps G, Dousset C, Maïga S, Derrien J, Thirouard L, Bouard L, Asnagli H, Beer P, Parker A, Gomez‐Bougie P, Devilder M, Moreau P, Touzeau C, Moreau‐Aubry A, Chiron D, Pellat‐Deceunynck C. Combined inhibition of CTPS1 and ATR is a metabolic vulnerability in p53-deficient myeloma cells. Hemasphere 2024; 8:e70016. [PMID: 39380841 PMCID: PMC11460984 DOI: 10.1002/hem3.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/12/2024] [Accepted: 07/31/2024] [Indexed: 10/10/2024] Open
Abstract
In multiple myeloma, as in B-cell malignancies, mono- and especially bi-allelic TP53 gene inactivation is a high-risk factor for treatment resistance, and there are currently no therapies specifically targeting p53 deficiency. In this study, we evaluated if the loss of cell cycle control in p53-deficient myeloma cells would confer a metabolically actionable vulnerability. We show that CTP synthase 1 (CTPS1), which encodes a CTP synthesis rate-limiting enzyme essential for DNA and RNA synthesis in lymphoid cells, is overexpressed in samples from myeloma patients displaying a high proliferation rate (high MKI67 expression) or a low p53 score (synonymous with TP53 deletion and/or mutation). This overexpression of CTPS1 was associated with reduced survival in two cohorts. Using scRNA-seq analysis in 24 patient samples, we further demonstrate that myeloma cells in the S or G2/M phase display high CTPS1 expression. Pharmacological inhibition of CTPS1 by STP-B induced cell cycle arrest in early S phase in isogenic NCI-H929 or XG7 TP53 +/+, TP53 -/-, and TP53 R175H/R175H cells and in a TP53 -/R123STOP patient sample. The functional annotation of transcriptional changes in 10 STP-B-treated myeloma cell lines revealed a decrease in protein translation and confirmed the blockade of cells into the S phase. The pharmacological inhibition of ATR, which governs the intrinsic S/G2 checkpoint, in STP-B-induced S-phase arrested cells synergistically induced cell death in TP53 +/+, TP53 -/-, and TP53 R175H/R175H isogenic cell lines (Bliss score >15). This combination induced replicative stress and caspase-mediated cell death and was highly effective in resistant/refractory patient samples with TP53 deletion and/or mutation and in TP53 -/- NCI-H929 xenografted NOD-scid IL2Rgamma mice. Our in vitro, ex vivo, and in vivo data provide the rationale for combined CTPS1 and ATR inhibition for the treatment of p53-deficient patients.
Collapse
Affiliation(s)
- Romane Durand
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Céline Bellanger
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Géraldine Descamps
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Christelle Dousset
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Sophie Maïga
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Jennifer Derrien
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Laura Thirouard
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Louise Bouard
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | | | | | | | | | | | - Philippe Moreau
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Cyrille Touzeau
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - Agnès Moreau‐Aubry
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | - David Chiron
- Nantes Université, INSERM, CHU NantesCNRS, Université d'Angers, CRCI2NANantesFrance
| | | |
Collapse
|
4
|
Durand R, Descamps G, Bellanger C, Dousset C, Maïga S, Alberge JB, Derrien J, Cruard J, Minvielle S, Lilli NL, Godon C, Le Bris Y, Tessoulin B, Amiot M, Gomez-Bougie P, Touzeau C, Moreau P, Chiron D, Moreau-Aubry A, Pellat-Deceunynck C. A p53 score derived from TP53 CRISPR/Cas9 HMCLs predicts survival and reveals a major role of BAX in the response to BH3 mimetics. Blood 2024; 143:1242-1258. [PMID: 38096363 DOI: 10.1182/blood.2023021581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/27/2023] [Accepted: 11/29/2023] [Indexed: 03/25/2024] Open
Abstract
ABSTRACT To establish a strict p53-dependent gene-expression profile, TP53-/- clones were derived from TP53+/+ and TP53-/mut t(4;14) human myeloma cell lines (HMCLs) using CRISPR/Cas9 technology. From the 17 dysregulated genes shared between the TP53-/- clones from TP53+/+ HMCLs, we established a functional p53 score, involving 13 genes specifically downregulated upon p53 silencing. This functional score segregated clones and myeloma cell lines as well as other cancer cell lines according to their TP53 status. The score efficiently identified samples from patients with myeloma with biallelic TP53 inactivation and was predictive of overall survival in Multiple Myeloma Research Foundation-coMMpass and CASSIOPEA cohorts. At the functional level, we showed that among the 13 genes, p53-regulated BAX expression correlated with and directly affected the MCL1 BH3 mimetic S63845 sensitivity of myeloma cells by decreasing MCL1-BAX complexes. However, resistance to S63845 was overcome by combining MCL1 and BCL2 BH3 mimetics, which displayed synergistic efficacy. The combination of BH3 mimetics was effective in 97% of patient samples with or without del17p. Nevertheless, single-cell RNA sequencing analysis showed that myeloma cells surviving the combination had lower p53 score, showing that myeloma cells with higher p53 score were more sensitive to BH3 mimetics. Taken together, we established a functional p53 score that identifies myeloma cells with biallelic TP53 invalidation, demonstrated that p53-regulated BAX is critical for optimal cell response to BH3 mimetics, and showed that MCL1 and BCL2 BH3 mimetics in combination may be of greater effectiveness for patients with biallelic TP53 invalidation, for whom there is still an unmet medical need.
Collapse
Affiliation(s)
- Romane Durand
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Géraldine Descamps
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Céline Bellanger
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Christelle Dousset
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - Sophie Maïga
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - Jean-Baptiste Alberge
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Jennifer Derrien
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Jonathan Cruard
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Stéphane Minvielle
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | | | | | - Yannick Le Bris
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - Benoit Tessoulin
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - Martine Amiot
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Patricia Gomez-Bougie
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - Cyrille Touzeau
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - Philippe Moreau
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
- Hematology Department, Nantes Université, Centre Hospitalier Universitaire (CHU) de Nantes, CNRS, INSERM, CRCI2NA, Nantes, France
| | - David Chiron
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Agnès Moreau-Aubry
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| | - Catherine Pellat-Deceunynck
- Molecular Vulnerabilities of Tumor Escape in B-cell Malignancies, Nantes Université, Centre National de la Recherche Scientifique (CNRS), INSERM, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes Angers (CRCI2NA), Nantes, France
- Hematology Department, Site de Recherche Intégrée sur le Cancer, Imaging and Longitudinal Investigations to Ameliorate Decision-making (SIRIC ILIAD), Nantes, France
| |
Collapse
|
5
|
Palanichamy JK, Tran TM, King JK, Katzman S, Ritter AJ, Sharma G, Tso C, Contreras JR, Fernando TR, Sanford JR, Rao DS. Distinct oncogenic phenotypes in hematopoietic specific deletions of Trp53. Sci Rep 2023; 13:7490. [PMID: 37160922 PMCID: PMC10169790 DOI: 10.1038/s41598-023-33949-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
Loss of function in the tumor suppressor gene TP53 is the most common alteration seen in human cancer. In mice, P53 deletion in all cells leads predominantly to the development of T-cell lymphomas, followed by B-cell lymphomas, sarcomas and teratomas. In order to dissect the role of P53 in the hematopoietic system, we generated and analyzed two different mouse models deficient for P53. A pan-hematopoietic P53 deletion mouse was created using Vav1-Cre based deletion; and a B-cell-specific deletion mouse was created using a CD19-Cre based deletion. The Vav1-P53CKO mice predominantly developed T-cell malignancies in younger mice, and myeloid malignancies in older mice. In T-cell malignancies, there was accelerated thymic cell maturation with overexpression of Notch1 and its downstream effectors. CD19-P53CKO mice developed marginal zone expansion in the spleen, followed by marginal zone lymphoma, some of which progressed to diffuse large B-cell lymphomas. Interestingly, marginal zone and diffuse large B-cell lymphomas had a unique gene expression signature characterized by activation of the PI3K pathway, compared with wild type marginal zone or follicular cells of the spleen. This study demonstrates lineage specific P53 deletion leading to distinct phenotypes secondary to unique gene expression programs set in motion.
Collapse
Affiliation(s)
| | - Tiffany M Tran
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
- Molecular, Cellular and Integrative Physiology Graduate Program, UCLA, Los Angeles, USA
| | | | - Sol Katzman
- Center for Biomolecular Science & Engineering, UCSC, Santa Cruz, USA
| | - Alexander J Ritter
- Department of Molecular, Cell and Developmental Biology and Center for Molecular Biology of RNA, UCSC, Santa Cruz, USA
| | - Gunjan Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Christine Tso
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | - Jorge R Contreras
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA
| | | | - Jeremy R Sanford
- Department of Molecular, Cell and Developmental Biology and Center for Molecular Biology of RNA, UCSC, Santa Cruz, USA
| | - Dinesh S Rao
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, USA.
- Broad Stem Cell Research Center, UCLA, Los Angeles, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 650 Charles E Young Drive, Los Angeles, CA, 90095, USA.
| |
Collapse
|
6
|
Webb F, Morey A, Mahler-Hinder C, Georgousopoulou E, Koo R, Pati N, Talaulikar D. Comprehensive FISH testing using FFPE tissue microarray of primary lymph node tissue identifies secondary cytogenetic abnormalities in Mantle Cell Lymphoma. Cancer Genet 2023; 274-275:75-83. [PMID: 37094546 DOI: 10.1016/j.cancergen.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 04/07/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023]
Abstract
INTRODUCTION Mantle Cell Lymphoma (MCL), is characterised by the reciprocal translocation t(11;14) resulting in CCND1-IGH gene fusion and subsequent upregulation of the CCND1 gene. Rearrangements of MYC and losses of CDKN2A and TP53 have been identified as biomarkers informing prognostic and potentially therapeutic information however these are not routinely assessed in MCL investigation. We aimed to identify additional cytogenetic changes using fluorescence in situ hybridisation (FISH) on formalin fixed paraffin embedded (FFPE) primary lymph node tissue microarrays in a cohort of 28 patients diagnosed with MCL between 2004 and 2019. FISH results were compared with corresponding immunohistochemistry (IHC) biomarkers to determine if IHC was a reliable screening tool to direct FISH testing. METHOD FFPE lymph node tissue samples were constructed into tissue microarrays (TMA) which were stained with 7 immunohistochemical biomarkers: Cyclin D1, c-Myc, p16, ATM, p53, Bcl-6 and Bcl-2. The same TMAs were hybridised with FISH probes for the corresponding genes; CCND1-IGH, MYC, CDKN2A, ATM, TP53, BCL6 and BCL2. FISH and the corresponding IHC biomarkers were analysed to determine if secondary cytogenetic changes could be identified and if IHC could be used as a reliable, inexpensive predictor of FISH abnormalities to potentially direct FISH testing. RESULTS CCND1-IGH fusion was detected in 27/28 (96%) of samples. Additional cytogenetic changes were identified by FISH in 15/28 (54%) of samples. Two additional abnormalities were detected in 2/28 (7%) samples. Cyclin D1 IHC overexpression was an excellent predictor of CCND1-IGH fusion. MYC and ATM IHC were useful screening tests to direct FISH testing and identified cases with poor prognostic features including blastoid change. IHC did not show clear concordance with FISH for other biomarkers. CONCLUSION FISH using FFPE primary lymph node tissue can detect secondary cytogenetic abnormalities in patients with MCL which are associated with an inferior prognosis. An expanded FISH panel including MYC, CDKN2A, TP53 and ATM should be considered in cases where anomalous IHC expression or is seen for these markers or if the patient appears to have the blastoid variant of the disease.
Collapse
Affiliation(s)
- Fiona Webb
- Department of Diagnostic Genomics, ACT Pathology, Canberra Health Services, Canberra, Australia.
| | - Adrienne Morey
- Department of Anatomical Pathology, ACT Pathology, Canberra Health Services, Canberra, Australia; Australian National University, Canberra, Australia
| | | | | | - RayMun Koo
- Haematology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Nalini Pati
- Department of Haematology, ACT Pathology, Canberra Health Services, Canberra, Australia
| | - Dipti Talaulikar
- Department of Diagnostic Genomics, ACT Pathology, Canberra Health Services, Canberra, Australia; Australian National University, Canberra, Australia; Department of Haematology, ACT Pathology, Canberra Health Services, Canberra, Australia
| |
Collapse
|
7
|
Recombinant human p53 adenovirus injection combined with Bortezomib inhibits proliferation and promotes apoptosis in multiple myeloma. Leuk Res 2023; 127:107041. [PMID: 36801701 DOI: 10.1016/j.leukres.2023.107041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Multiple myeloma (MM) is a B-cell malignancy characterized by abnormal proliferation of clonal plasma cells in the bone marrow, the incidence of which has further increased in recent years. In multiple myeloma, wild-type functional p53 is often inactivated or dysregulated. Therefore, this study aimed to investigate the role of p53 knockdown or overexpression in multiple myeloma and the therapeutic effect of recombinant adenovirus-p53 (rAd-p53) in combination with Bortezomib. METHODS SiRNA p53 and rAd-p53 were used to knock down and overexpress p53. RT-qPCR was used to detect gene expression, and western blotting (WB) was used to detect protein expression levels. We also constructed wild-type multiple myeloma cell line-MM1S cell xenograft tumor models and explored the effects of siRNA-p53, rAd-p53, and Bortezomib on multiple myeloma in vivo and in vitro. H&E staining and KI67 immunohistochemical staining were used to assess the anti-myeloma effects of recombinant adenovirus and Bortezomib in vivo. RESULTS The designed siRNA p53 effectively led to the knockdown of the p53 gene, while rAd-p53 could significantly achieve p53 overexpression. p53 gene inhibited MM1S cell proliferation and promoted apoptosis of wild-type multiple myeloma cell line MM1S. P53 gene inhibited tumor proliferation in vitro by promoting p21 expression and reducing cell cycle protein B1 expression of MM1S. P53 gene overexpression could inhibit tumor growth in vivo. Injection of rAd-p53 in tumor models inhibited tumor development through p21- and cyclin B1-mediated cell proliferation and apoptosis regulation. CONCLUSIONS We found that overexpression of p53 inhibits MM tumor cell survival and proliferation in vivo and in vitro. Furthermore, the combination of rAd-p53 and Bortezomib significantly improved the efficacy, which provides a new possibility for more effective treatment of MM.
Collapse
|
8
|
Li C, Zhang Y, Xiao Y, Luo Y. Identifying the Effect of COVID-19 Infection in Multiple Myeloma and Diffuse Large B-Cell Lymphoma Patients Using Bioinformatics and System Biology. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7017317. [PMID: 36466549 PMCID: PMC9711963 DOI: 10.1155/2022/7017317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/05/2022] [Accepted: 11/12/2022] [Indexed: 09/29/2023]
Abstract
The severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), also referred to as COVID-19, has spread to several countries and caused a serious threat to human health worldwide. Patients with confirmed COVID-19 infection spread the disease rapidly throughout the region. Multiple myeloma (MM) and diffuse large B-cell lymphoma (DLBCL) are risk factors for COVID-19, although the molecular mechanisms underlying the relationship among MM, DLBCL, and COVID-19 have not been elucidated so far. In this context, transcriptome analysis was performed in the present study to identify the shared pathways and molecular indicators of MM, DLBCL, and COVID-19, which benefited the overall understanding of the effect of COVID-19 in patients with MM and DLBCL. Three datasets (GSE16558, GSE56315, and GSE152418) were downloaded from the Gene Expression Omnibus (GEO) and searched for the shared differentially expressed genes (DEGs) in patients with MM and DLBCL who were infected with SARS-CoV-2. The objective was to detect similar pathways and prospective medicines. A total of 29 DEGs that were common across these three datasets were selected. A protein-protein interaction (PPI) network was constructed using data from the STRING database followed by the identification of hub genes. In addition, the association of MM and DLBCL with COVID-19 infection was analyzed through functional analysis using ontologies terms and pathway analysis. Three relationships were observed in the evaluated datasets: transcription factor-gene interactions, protein-drug interactions, and an integrated regulatory network of DEGs and miRNAs with mutual DEGs. The findings of the present study revealed potential pharmaceuticals that could be beneficial in the treatment of COVID-19.
Collapse
Affiliation(s)
- Chengcheng Li
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ying Zhang
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingying Xiao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Yun Luo
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Cui Y, Wen Y, Lv C, Zhao D, Yang Y, Qiu H, Wang C. Decreased RNA‑binding protein IGF2BP2 downregulates NT5DC2, which suppresses cell proliferation, and induces cell cycle arrest and apoptosis in diffuse large B‑cell lymphoma cells by regulating the p53 signaling pathway. Mol Med Rep 2022; 26:286. [PMID: 35894142 PMCID: PMC9366151 DOI: 10.3892/mmr.2022.12802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/23/2022] [Indexed: 11/06/2022] Open
Abstract
Diffuse large B‑cell lymphoma (DLBCL) remains difficult to treat clinically due to its highly aggressive characteristics. Insulin‑like growth factor 2 mRNA‑binding protein 2 (IGF2BP2) and 5'‑nucleotidase domain‑containing 2 (NT5DC2) have been suggested as potential regulators in numerous types of cancer. The present study aimed to determine whether downregulation of IGF2BP2 and NT5DC2 suppresses cell proliferation, and induces cell cycle arrest and apoptosis in DLBCL cells by regulating the p53 signaling pathway. The expression levels of IGF2BP2 and NT5DC2 in DLBCL cells were determined by reverse transcription‑quantitative PCR (RT‑qPCR) and western blot analysis. Transfection of cells with IGF2BP2 overexpressing plasmids and NT5DC2 interference plasmids was performed, and the efficacy of transfection was confirmed by RT‑qPCR and western blot analysis. The viability, proliferation, cell cycle progression and apoptosis of DLBCL cells were analyzed by Cell Counting Kit‑8 assay, 5‑bromo‑2‑deoxyuridine staining and flow cytometry. RNA pull‑down and immunoprecipitation assays were used to verify the binding of IGF2BP2 and NT5DC2. The expression levels of apoptosis, cell cycle and p53 pathway‑associated proteins were determined by western blotting. The results revealed that NT5DC2 expression was increased in DLBCL cell lines and was the highest in OCI‑Ly7 cells. IGF2BP2 expression was also increased in OCI‑Ly7 cells and IGF2BP2 bound to NT5DC2. Knockdown of NT5DC2 suppressed cell viability and proliferation, induced cell cycle arrest and promoted apoptosis in DLBCL cells, which was reversed by upregulation of IGF2BP2. In addition, knockdown of NT5DC2 increased the expression of p53 and p21, but suppressed the expression of proliferating cell nuclear antigen, CDK4 and cyclin D1; these effects were reversed by upregulation of IGF2BP2. In conclusion, knockdown of NT5DC2 suppressed cell viability and proliferation, induced cell cycle arrest and promoted apoptosis in DLBCL cells by regulating the p53 signaling pathway and these effects were reversed by upregulation of IGF2BP2.
Collapse
Affiliation(s)
- Yuying Cui
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Yu Wen
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Chao Lv
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Dongmei Zhao
- School of Clinical Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Yu Yang
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China
| | - Hongbin Qiu
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China,School of Public Health, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China,Correspondence to: Dr Chennan Wang or Dr Hongbin Qiu, School of Basic Medicine, Jiamusi University, 148 Xuefu Street, Jiamusi, Heilongjiang 154007, P.R. China, E-mail: , E-mail:
| | - Chennan Wang
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang 154007, P.R. China,Correspondence to: Dr Chennan Wang or Dr Hongbin Qiu, School of Basic Medicine, Jiamusi University, 148 Xuefu Street, Jiamusi, Heilongjiang 154007, P.R. China, E-mail: , E-mail:
| |
Collapse
|
10
|
Zhang X, Wu Z, Hao Y, Yu T, Li X, Liang Y, Li J, Huang L, Xu Y, Li X, Xu X, Wang W, Xu G, Zhang X, Lv Q, Fang Y, Xu R, Qian W. Aberrantly Activated APOBEC3B Is Associated With Mutant p53-Driven Refractory/Relapsed Diffuse Large B-Cell Lymphoma. Front Immunol 2022; 13:888250. [PMID: 35592333 PMCID: PMC9112561 DOI: 10.3389/fimmu.2022.888250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor protein 53 (TP53) mutation predicts an unfavorable prognosis in diffuse large B-cell lymphoma (DLBCL), but the molecular basis for this association remains unclear. In several malignancies, the cytidine deaminase apolipoprotein B mRNA editing enzyme catalytic subunit 3B (APOBEC3B) has been reported to be associated with the TP53 G/C-to-A/T mutation. Here, we show that the frequency of this mutation was significantly higher in relapsed/refractory (R/R) than in non-R/R DLBCL, which was positively associated with the APOBEC3B expression level. APOBEC3B overexpression induced the TP53 G/C-to-A/T mutation in vitro, resulting in a phenotype similar to that of DLBCL specimens. Additionally, APOBEC3B-induced p53 mutants promoted the growth of DLBCL cells and enhanced drug resistance. These results suggest that APOBEC3B is a critical factor in mutant p53-driven R/R DLBCL and is therefore a potential therapeutic target.
Collapse
Affiliation(s)
- Xuzhao Zhang
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Zhejiang University, Hangzhou, China
| | - Zhaoxing Wu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanyuan Hao
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Teng Yu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xian Li
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Liang
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jinfan Li
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Liansheng Huang
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Xu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuzhen Li
- Department of Pathology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaohua Xu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiqin Wang
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Genbo Xu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohong Zhang
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qinghua Lv
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yongming Fang
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Rongzhen Xu
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
11
|
Cencini E, Fabbri A, Raspadori D, Gozzetti A, Bocchia M. Tp53 disruptions: is there a marker of poor prognosis in chronic lymphoproliferative disorders? Blood Res 2021; 56:333-334. [PMID: 34743076 PMCID: PMC8721443 DOI: 10.5045/br.2021.2020322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 07/06/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Affiliation(s)
- Emanuele Cencini
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| | - Alberto Fabbri
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| | - Donatella Raspadori
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| | - Alessandro Gozzetti
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| | - Monica Bocchia
- Unit of Hematology, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| |
Collapse
|
12
|
Dang W, Cao P, Yan Q, Yang L, Wang Y, Yang J, Xin S, Zhang J, Li J, Long S, Zhang W, Zhang S, Lu J. IGFBP7-AS1 is a p53-responsive long noncoding RNA downregulated by Epstein-Barr virus that contributes to viral tumorigenesis. Cancer Lett 2021; 523:135-147. [PMID: 34634383 DOI: 10.1016/j.canlet.2021.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/16/2021] [Accepted: 10/05/2021] [Indexed: 01/15/2023]
Abstract
Epstein-Barr virus (EBV) is closely related to the development of several malignancies, such as B-cell lymphoma (B-CL), by the mechanism through which these malignancies develop remains largely unknown. We previously observed downregulation of the long noncoding RNA (lncRNA) IGFBP7-AS1 in response to EBV infection. However, the role of IGFBP7-AS1 in EBV-associated cancers has not been clarified. Here, we found that expression of IGFBP7-AS1, as well as its sense gene IGFBP7, is decreased in EBV-positive B-CL cells and clinical tissues. IGFBP7-AS1 stabilizes IGFBP7 mRNA by forming a duplex based on their overlapping regions. The tumour suppressor p53 transcriptionally activates IGFBP7-AS1 expression by binding to the promoter region of the lncRNA gene. The IGFBP7-AS1 expression is able to be rescued in EBV-positive cells in wild-type (wt) p53-dependent manner. IGFBP7-AS1 inhibits the proliferation and promotes the apoptosis of B-CL cells. Moreover, tumorigenic properties due to the depletion of IGFBP7-AS1 were restored by exogenous expression of IGFBP7 or wt-p53. Furthermore, the functional p53/IGFBP7-AS1/IGFBP7 axis facilitates apoptosis by suppressing the production and secretion of the NPPB signal peptide and further regulating the cGMP-PKG signalling pathway. This study demonstrates that EBV promotes tumorigenesis, particularly in B-CL progression, by downregulating the novel p53-responsive lncRNA IGFBP7-AS1.
Collapse
Affiliation(s)
- Wei Dang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Li Yang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Yiwei Wang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Jing Yang
- NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Shuyu Xin
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Jing Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Jing Li
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Sijing Long
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Wentao Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Senmiao Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China
| | - Jianhong Lu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, 410078, Hunan, China; NHC Key Laboratory of Carcinogenesis, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, 410078, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410080, Hunan, China; China-Africa Research Center of Infectious Diseases, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
13
|
Aksenova AY, Zhuk AS, Lada AG, Zotova IV, Stepchenkova EI, Kostroma II, Gritsaev SV, Pavlov YI. Genome Instability in Multiple Myeloma: Facts and Factors. Cancers (Basel) 2021; 13:5949. [PMID: 34885058 PMCID: PMC8656811 DOI: 10.3390/cancers13235949] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a malignant neoplasm of terminally differentiated immunoglobulin-producing B lymphocytes called plasma cells. MM is the second most common hematologic malignancy, and it poses a heavy economic and social burden because it remains incurable and confers a profound disability to patients. Despite current progress in MM treatment, the disease invariably recurs, even after the transplantation of autologous hematopoietic stem cells (ASCT). Biological processes leading to a pathological myeloma clone and the mechanisms of further evolution of the disease are far from complete understanding. Genetically, MM is a complex disease that demonstrates a high level of heterogeneity. Myeloma genomes carry numerous genetic changes, including structural genome variations and chromosomal gains and losses, and these changes occur in combinations with point mutations affecting various cellular pathways, including genome maintenance. MM genome instability in its extreme is manifested in mutation kataegis and complex genomic rearrangements: chromothripsis, templated insertions, and chromoplexy. Chemotherapeutic agents used to treat MM add another level of complexity because many of them exacerbate genome instability. Genome abnormalities are driver events and deciphering their mechanisms will help understand the causes of MM and play a pivotal role in developing new therapies.
Collapse
Affiliation(s)
- Anna Y. Aksenova
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anna S. Zhuk
- International Laboratory “Computer Technologies”, ITMO University, 197101 St. Petersburg, Russia;
| | - Artem G. Lada
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA;
| | - Irina V. Zotova
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (I.V.Z.); (E.I.S.)
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, 199034 St. Petersburg, Russia
| | - Elena I. Stepchenkova
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (I.V.Z.); (E.I.S.)
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, 199034 St. Petersburg, Russia
| | - Ivan I. Kostroma
- Russian Research Institute of Hematology and Transfusiology, 191024 St. Petersburg, Russia; (I.I.K.); (S.V.G.)
| | - Sergey V. Gritsaev
- Russian Research Institute of Hematology and Transfusiology, 191024 St. Petersburg, Russia; (I.I.K.); (S.V.G.)
| | - Youri I. Pavlov
- Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Departments of Biochemistry and Molecular Biology, Microbiology and Pathology, Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
14
|
The European Medicines Agency Review of Tafasitamab in Combination With Lenalidomide for the Treatment of Adult Patients With Relapsed/Refractory Diffuse Large B-cell Lymphoma. Hemasphere 2021; 5:e666. [PMID: 34805769 PMCID: PMC8601272 DOI: 10.1097/hs9.0000000000000666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/01/2021] [Indexed: 11/26/2022] Open
Abstract
Tafasitamab is a humanized monoclonal antibody that binds to the CD19 antigen, which is expressed in tumor cells from patients with diffuse large B-cell lymphoma (DLBCL). On June 24, 2021, a positive opinion for a conditional marketing authorization was issued by the European Medicines Agency (EMA)’s Committee for Medicinal Products for Human Use (CHMP) for tafasitamab, in combination with lenalidomide, for the treatment of adult patients with relapsed or refractory DLBCL who are ineligible for autologous stem cell transplantation. Tafasitamab was evaluated in the phase 2 single-arm, multicenter, open-label L-MIND clinical trial. The primary endpoint of this trial was objective response rate (ORR). The best ORR, achieved at any time during the study, was 56.8% (95% confidence interval: 45.3%–67.8%), and the median duration of response was 34.6 months (95% confidence interval: 26.1–not reached). The most frequently reported adverse events by system organ class were infections and infestations (72.8%; grade ≥3: 29.6%), blood and lymphatic system disorders (65.4%; grade ≥3: 56.8%), gastrointestinal disorders (64.2%; grade ≥3: 2.5%), and general disorders and administration site conditions (58.0%; grade ≥3: 8.6%). The aim of this article is to summarize the scientific review of the application which led to the positive opinion by the CHMP.
Collapse
|
15
|
Ku J, Kim R, Kim D, Kim D, Song S, Lee K, Lee N, Kim M, Yoon SS, Kwon NH, Kim S, Kim Y, Koh Y. Single-cell analysis of AIMP2 splice variants informs on drug sensitivity and prognosis in hematologic cancer. Commun Biol 2020; 3:630. [PMID: 33128014 PMCID: PMC7599330 DOI: 10.1038/s42003-020-01353-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 09/22/2020] [Indexed: 11/21/2022] Open
Abstract
Aminoacyl-tRNA synthetase-interacting multifunctional protein 2 (AIMP2) is a non-enzymatic component required for the multi-tRNA synthetase complex. While exon 2 skipping alternatively spliced variant of AIMP2 (AIMP2-DX2) compromises AIMP2 activity and is associated with carcinogenesis, its clinical potential awaits further validation. Here, we found that AIMP2-DX2/AIMP2 expression ratio is strongly correlated with major cancer signaling pathways and poor prognosis, particularly in acute myeloid leukemia (AML). Analysis of a clinical patient cohort revealed that AIMP2-DX2 positive AML patients show decreased overall survival and progression-free survival. We also developed targeted RNA-sequencing and single-molecule RNA-FISH tools to quantitatively analyze AIMP2-DX2/AIMP2 ratios at the single-cell level. By subclassifying hematologic cancer cells based on their AIMP2-DX2/AIMP2 ratios, we found that downregulating AIMP2-DX2 sensitizes cells to anticancer drugs only for a subgroup of cells while it has adverse effects on others. Collectively, our study establishes AIMP2-DX2 as a potential biomarker and a therapeutic target for hematologic cancer. Ku, Kim et al develop a method to analyse the ratio of the alternatively spliced variant of AIMP2 to full length AIMP via single-molecule RNA-FISH. They can subclassify hematologic cancer based on AIMP2-DX2/AIMP2 ratio and find that cells with high AIMP2-DX2 ratio can be sensitized to chemotherapy drugs by depleting AIMP2-DX2.
Collapse
Affiliation(s)
- Jayoung Ku
- Department of Chemical and Biomolecular Engineering and KAIST Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ryul Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dongchan Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Daeyoon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seulki Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Keonyong Lee
- Department of Chemical and Biomolecular Engineering and KAIST Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Namseok Lee
- Department of Chemical and Biomolecular Engineering and KAIST Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - MinA Kim
- Department of Chemical and Biomolecular Engineering and KAIST Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Nam Hoon Kwon
- Medicinal Bioconvergence Research Center, Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sunghoon Kim
- Medicinal Bioconvergence Research Center, Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Yoosik Kim
- Department of Chemical and Biomolecular Engineering and KAIST Institute for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| | - Youngil Koh
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea. .,Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
16
|
ROS Overproduction Sensitises Myeloma Cells to Bortezomib-Induced Apoptosis and Alleviates Tumour Microenvironment-Mediated Cell Resistance. Cells 2020; 9:cells9112357. [PMID: 33114738 PMCID: PMC7693395 DOI: 10.3390/cells9112357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm that remains incurable due to innate or acquired resistance. Although MM cells produce high intracellular levels of reactive oxygen species (ROS), we hypothesised that they could remain sensitive to ROS unbalance. We tested if the inhibition of ROS, on one hand, or the overproduction of ROS, on the other, could (re)sensitise cells to bortezomib (BTZ). Two drugs were used in a panel of MM cell lines with various responses to BTZ: VAS3947 (VAS), an inhibitor of NADPH oxidase and auranofin (AUR), an inhibitor of thioredoxin reductase (TXNRD1), an antioxidant enzyme overexpressed in MM cells. We used several culture models: in suspension, on a fibronectin layer, in coculture with HS-5 mesenchymal cells, and/or in 3-D culture (or spheroids) to study the response of MM primary cells and cell lines. Several MM cell lines were sensitive to VAS but the combination with BTZ showed antagonistic or additive effects at best. By contrast, in all culture systems studied, the combined AUR/BTZ treatment showed synergistic effects on cell lines, including those less sensitive to BTZ and primary cells. MM cell death is due to the activation of apoptosis and autophagy. Modulating the redox balance of MM cells could be an effective therapy for refractory or relapse post-BTZ patients.
Collapse
|
17
|
Le Bris Y, Magrangeas F, Moreau A, Chiron D, Guérin-Charbonnel C, Theisen O, Pichon O, Canioni D, Burroni B, Maisonneuve H, Thieblemont C, Oberic L, Gyan E, Pellat-Deceunynck C, Hermine O, Delfau-Larue MH, Tessoulin B, Béné MC, Minvielle S, Le Gouill S. Whole genome copy number analysis in search of new prognostic biomarkers in first line treatment of mantle cell lymphoma. A study by the LYSA group. Hematol Oncol 2020; 38:446-455. [PMID: 32472610 DOI: 10.1002/hon.2750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/11/2020] [Accepted: 05/24/2020] [Indexed: 11/09/2022]
Abstract
Mantle cell lymphoma (MCL) is a lymphoproliferative disorder characterized by the t(11;14)(q13;q32) CCND1/IGH translocation. This lymphoma is however extremely heterogeneous in terms of molecular alterations. Moreover, the course of the disease can vary greatly between indolent forms with slow progression and aggressive conditions rapidly pejorative. The identification of early markers allowing to predict individual patients outcome has however been unsuccessful so far. The LyMa trial treated homogeneously a cohort of young MCL patients. This appeared as a good opportunity to search for biomarkers of response to therapy. DNA extracted from diagnostic paraffin-embedded lymph node biopsies from 100 patients with newly diagnosed MCL, homogeneously treated in this prospective clinical trial, were investigated for copy number alterations and copy neutral loss of heterozygosity using the Oncoscan SNP-array scanning the whole genome. An independent confirmatory cohort was used to strengthen the possibly relevant anomalies observed. Here we describe the recurrent anomalies identified with this technique. Deletions of 17p(TP53) and 9p(CDKN2A) were more frequent in refractory or early relapsing patients (10%), but had no significant impact in univariate analysis on progression-free (PFS) or overall survival (OS). Regardless of the presence of TP53 or CDKN2A deletions, gains in 7p22 (8,5%) were associated with better PFS in univariate but not in multivariate analysis including MCL International Prognostic Index and treatment. Gains of 11q(CCDN1), suggesting gains of the CCND1/IGH fusion, were associated with worse OS and PFS in univariate and multivariate analyses. This worse prognosis impact was confirmed by FISH in an independent confirmatory cohort. This work, using a whole genome approach, confirms the broad genomic landscape of MCL and shows that gains of the CCND1/IGH fusion can be considered as a new prognostic structural variant. Genomic abnormalities of prognostic impact could be useful to strengthen or de-escalate treatment schedules or choosing targeted therapies or CART-cells.
Collapse
Affiliation(s)
- Yannick Le Bris
- Hematology Biology Department, Nantes University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Florence Magrangeas
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Anne Moreau
- Pathology Department Nantes University Hospital, now in Centre Hospitalier Départemental de Vendée, La Roche sur Yon, France
| | - David Chiron
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Catherine Guérin-Charbonnel
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Institut de Cancérologie de l'Ouest, U892, Saint-Herblain, France
| | - Olivier Theisen
- Hematology Biology Department, Nantes University Hospital, Nantes, France
| | - Olivier Pichon
- Genetic Department, Nantes University Hospital, Nantes, France
| | | | - Barbara Burroni
- Pathology Department, Cochin University Hospital, Paris, France
| | - Hervé Maisonneuve
- Hematology Clinic, Centre Hospitalier Départemental de Vendée, La Roche sur Yon, France
| | | | - Lucie Oberic
- Clinical Hematology Department, IUCT Oncopole, Toulouse University Hospital, Toulouse, France
| | - Emmanuel Gyan
- Clinical Hematology Department, Tours University Hospital, Tours, France
| | | | - Olivier Hermine
- Clinical Hematology Department, Necker University Hospital, Paris, France
| | | | - Benoît Tessoulin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Clinical Hematology Department, Nantes University Hospital, Nantes, France
| | - Marie-Christine Béné
- Hematology Biology Department, Nantes University Hospital, Nantes, France.,CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Stéphane Minvielle
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Steven Le Gouill
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Clinical Hematology Department, Nantes University Hospital, Nantes, France
| |
Collapse
|
18
|
The prognostic importance of double-expressor subgroup and AID , UNG and mismatch repair protein expressions in diffuse large B-cell lymphomas. MARMARA MEDICAL JOURNAL 2020. [DOI: 10.5472/marumj.741651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
19
|
Mirza AS, Horna P, Teer JK, Song J, Akabari R, Hussaini M, Sokol L. New Insights Into the Complex Mutational Landscape of Sézary Syndrome. Front Oncol 2020; 10:514. [PMID: 32373524 PMCID: PMC7186303 DOI: 10.3389/fonc.2020.00514] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 03/23/2020] [Indexed: 12/28/2022] Open
Abstract
Sézary syndrome (SS) is a genetically and clinically distinct entity among cutaneous T-cell lymphomas (CTCL). SS is characterized by more aggressive disease compared to the most common indolent type of CTCL, mycosis fungoides. However, there are limited available genomic data regarding SS. To characterize and expand current mappings of the genomic landscape of CTCL, whole exome sequencing (WES) was performed on peripheral blood samples from seven patients with SS. We detected 21,784 variants, of which 21,140 were novel and 644 were previously described. Filtering revealed 551 nonsynonymous variants among 525 mutated genes−25 recurrent mutations and 1 recurrent variant. Several recurrently mutated genes crucial to pathogenesis pathways, including Janus kinase (JAK)/signal transducers and activators of transcription (STAT), peroxisome proliferator-activated receptors (PPAR), PI3K-serine/threonine protein kinases (AKT), and fibroblast growth factor receptors (FGFR), were identified. Furthermore, genetic mutations spanned both known and novel genes, supporting the idea of a long-tail distribution of mutations in lymphoma. Acknowledging these genetic variants and their affected pathways may inspire future targeted therapies. WES of a limited number of SS patients revealed both novel findings and corroborated complexities of the “long-tail” distribution of previously reported mutations.
Collapse
Affiliation(s)
- Abu-Sayeef Mirza
- Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Pedro Horna
- Division of Hematopathology, Mayo Clinic, Rochester, MN, United States
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Jinming Song
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Ratilal Akabari
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Mohammad Hussaini
- Department of Hematopathology and Laboratory Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Lubomir Sokol
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
20
|
Holthof LC, van der Horst HJ, van Hal-van Veen SE, Ruiter RWJ, Li F, Buijze M, Andersen MN, Yuan H, de Bruijn J, van de Donk NWCJ, Lokhorst HM, Zweegman S, Groen RWJ, Mutis T. Preclinical evidence for an effective therapeutic activity of FL118, a novel survivin inhibitor, in patients with relapsed/refractory multiple myeloma. Haematologica 2020; 105:e80-e83. [PMID: 31123033 DOI: 10.3324/haematol.2018.213314] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Lisa C Holthof
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Hilma J van der Horst
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Susan E van Hal-van Veen
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ruud W J Ruiter
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, New York, NJ USA
| | - Marijke Buijze
- Department of Otolaryngology-Head and Neck Surgery, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Morten N Andersen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Huipin Yuan
- Kuros Biosciences BV, Bilthoven, the Netherlands
| | - Joost de Bruijn
- Kuros Biosciences BV, Bilthoven, the Netherlands.,The School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Henk M Lokhorst
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Richard W J Groen
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Tuna Mutis
- Department of Hematology, Amsterdam UMC, VU University Medical Center, Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
21
|
Yu L, Yu TT, Young KH. Cross-talk between Myc and p53 in B-cell lymphomas. Chronic Dis Transl Med 2019; 5:139-154. [PMID: 31891126 PMCID: PMC6926120 DOI: 10.1016/j.cdtm.2019.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Indexed: 02/07/2023] Open
Abstract
Myc and p53 proteins are closely associated with many physiological cellular functions, including immune response and lymphocyte survival, and are expressed in the lymphoid organs, which are sites for the development and activation of B-cell malignancies. Genetic alterations and other mechanisms resulting in constitutive activation, rearrangement, or mutation of MYC and TP53 contribute to the development of lymphomas, progression and therapy resistance by gene dysregulation, activation of downstream anti-apoptotic pathways, and unfavorable microenvironment interactions. The cross-talk between the Myc and p53 proteins contributes to the inferior prognosis in many types of B-cell lymphomas. In this review, we present the physiological roles of Myc and p53 proteins, and recent advances in understanding the pathological roles of Myc, p53, and their cross-talk in lymphoid neoplasms. In addition, we highlight clinical trials of novel agents that directly or indirectly inhibit Myc and/or p53 protein functions and their signaling pathways. Although, to date, these trials have failed to overcome drug resistance, the new results have highlighted the clinical efficiency of targeting diverse mechanisms of action with the goal of optimizing novel therapeutic opportunities to eradicate lymphoma cells.
Collapse
Affiliation(s)
- Li Yu
- Department of Hematology, The Second Affiliated Hospital to Nanchang University, Nanchang, Jiangxi 330006, China
- Hematopathology Division and Pathology Department, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tian-Tian Yu
- Department of Hematology, The Second Affiliated Hospital to Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ken H. Young
- Hematopathology Division and Pathology Department, Duke University School of Medicine, Durham, NC 27710, USA
- Duke University Medical Center and Cancer Institute, Durham, NC 27710, USA
| |
Collapse
|
22
|
Tang C, Kuruvilla J. Optimal management of mantle cell lymphoma in the primary setting. Expert Rev Hematol 2019; 12:715-721. [PMID: 31268728 DOI: 10.1080/17474086.2019.1639501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: The management of mantle cell lymphoma (MCL) has significantly improved since the use of intensified induction and autologous stem cell transplant consolidation. Evolving developments in minimal residual disease detection and novel agent therapy are now challenging this frontline treatment paradigm. Areas covered: This review discusses both the established role of induction and transplant consolidation in MCL, followed by evolving concepts in the use of novel agents in the frontline setting, and the use of minimal residual disease as a driver of MCL management. Expert opinion: In an era of novel agents and improved biologic understanding of MCL, our goal for frontline management should evolve toward personalized therapy for individual patients to maximize efficacy and survival whilst minimizing treatment-related toxicities.
Collapse
Affiliation(s)
- Catherine Tang
- Division of Medical Oncology & Hematology, Princess Margaret Hospital , Toronto , Canada.,Department of Medicine, University of Toronto , Toronto , Canada
| | - John Kuruvilla
- Division of Medical Oncology & Hematology, Princess Margaret Hospital , Toronto , Canada.,Department of Medicine, University of Toronto , Toronto , Canada
| |
Collapse
|
23
|
Mitrakos A, Kattamis A, Katsibardi K, Papadhimitriou S, Kitsiou-Tzeli S, Kanavakis E, Tzetis M. High resolution Chromosomal Microarray Analysis (CMA) enhances the genetic profile of pediatric B-cell Acute Lymphoblastic Leukemia patients. Leuk Res 2019; 83:106177. [PMID: 31261022 DOI: 10.1016/j.leukres.2019.106177] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 11/26/2022]
Abstract
Acute Lymphoblastic Leukemia (ALL) is a malignancy of the immature lymphoid cells mainly associated with numerical and structural chromosomal aberrations. The current standard for profiling the diverse genetic background comprises a combination of conventional karyotype and FISH analysis for the most common translocations, albeit with many limitations. Chromosomal Microarray Analysis (CMA) is a high throughput whole genome method that is gradually implemented in routine clinical practice, but not many studies have compared the two methods. Here we aim to investigate the added benefits of utilizing the high resolution 2 x 400 K G3 CGH + SNP CMA platform in routine diagnostics of pediatric ALL. From the 29 bone marrow samples that were analyzed, CMA identified clinically relevant findings in 83%, while detecting chromosomal aberrations in 75% of the patients with normal conventional karyotype. The most common finding was hyperdiploidy (20%), and the most common submicroscopic aberration involved CDKN2A/B genes. The smallest aberration detected was a 9 kb partial NF1 gene duplication. The prognosis of the patients when combining conventional cytogenetics and CMA was either changed or enhanced in 66% of the cases. A rare duplication possibly indicative of a cryptic ABL1-NUP214 fusion gene was found in one patient. We conclude that CMA, when combined with conventional cytogenetic analysis, can significantly enhance the genetic profiling of patients with pediatric ALL in a routine clinical setting.
Collapse
Affiliation(s)
- Anastasios Mitrakos
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Antonis Kattamis
- Hematology-Oncology Unit, First Department of Pediatrics, National and Kapodistrian University of Athens, "Aghia Sofia" Children's Hospital, Athens, Greece
| | - Katerina Katsibardi
- Hematology-Oncology Unit, First Department of Pediatrics, National and Kapodistrian University of Athens, "Aghia Sofia" Children's Hospital, Athens, Greece
| | - Stefanos Papadhimitriou
- Department of Laboratory Hematology, Athens Regional General Hospital "G. Gennimatas", Athens, Greece
| | - Sophia Kitsiou-Tzeli
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Emmanuel Kanavakis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| | - Maria Tzetis
- Department of Medical Genetics, Medical School, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
24
|
Hu Y, Chen W, Wang J. Progress in the identification of gene mutations involved in multiple myeloma. Onco Targets Ther 2019; 12:4075-4080. [PMID: 31213829 PMCID: PMC6538831 DOI: 10.2147/ott.s205922] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/30/2019] [Indexed: 12/22/2022] Open
Abstract
Sequencing studies have been used to determine a spectrum of multiple myeloma (MM) mutations. Mutation of certain genes, including KRAS, NRAS, TP53, FAM46C, DIS3 and BRAF, have a high recurrence rate and may play important roles in the pathogenesis, progression and prognosis of MM. Mutations in DIS3, which encodes a highly conserved RNA exonuclease, lead to loss of function. The expression of FAM46C is highly correlated with the expression of ribosomal protein, but the exact function of FAM46C mutation is unclear. There are mutants of IRF4, which is considered an MM survival factor. Mutations in the gene coding for the DNA damage-binding protein (DDB1) may affect interactions with CUL4A, which is part of the cereblon (CRBN) ubiquitin ligase complex. IRF4is part of the complex, which binds to DNA. These findings might explain the resistance to immunomodulatory. TP53 deletion or mutation is often present in B-cell malignancies and is associated with low response rates. Myeloma pathogenic mutations in ATM have been found in adult lymphatic tumors. XBP1 and PSMB5 mutations may be related to bortezomib resistance. Multiple gene mutations (KRAS, NRAS and BRAF) involved in the same pathway were found a single patient. Identification of driver gene mutations has brought great hope to the field of individualized, targeted medicine for MM.
Collapse
Affiliation(s)
- Ying Hu
- Department of Hematology, Aerospace Central Hospital of Peking University, Beijing, People's Republic of China
| | - Wenming Chen
- Department of Hematology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jingbo Wang
- Department of Hematology, Aerospace Central Hospital of Peking University, Beijing, People's Republic of China
| |
Collapse
|
25
|
Shi K, Xia Y, Zhu HY, Wang L, Fan L, Xu W, Li JY. [High-dose methylprednisolone with Rituximab and fresh frozen plasma in the treatment of six patients with B-cell lymphoproliferative disorders harboring TP53 abnormalities]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:388-392. [PMID: 31207703 PMCID: PMC7342232 DOI: 10.3760/cma.j.issn.0253-2727.2019.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Indexed: 11/05/2022]
Abstract
Objective: To investigate whether high-dose methylprednisolone with Rituximab and fresh frozen plasma (HDMP+RTX+FFP) is an effective therapy for patients with B-cell chronic lymphoproliferative disorders (B-CLPD) with TP53 abnormalities. Methods: Six B-CLPD patients with TP53 abnormalities from May 2008 to May 2012 were prospectively enrolled in the study. The patients were treated with HDMP+RTX+FFP for up to 6 cycles. Results: Of the six B-CLPD patients, there were 4 cases of chronic B-cell lymphoproliferative disorders-unclassified (B-CLPD-U) , 1 B-cell prolymphocytic leukemia (B-PLL) and 1 mantle cell lymphoma (MCL) . After a median 3 courses of treatment, 4 patients achieved complete remission (CR) including 3 with undetectable minimal residual disease (MRD(-)) . One patient was evaluated as stable disease (SD) and another one patient was in disease progression (PD) . After a median follow-up of 30 (4-56) months, 2 non-responders progressed quickly and died. All of CR patients survived and no one succumbed to disease progression at the last follow-up. The hematopoietic function was significantly improved after the treatment whereas there was also significant decrease in serum IgA, IgG and IgM levels. All patients showed well tolerance to this regimen. The incidence of myelosuppression was low and adverse events (AE) were mainly neutropenia which did not exceed grade 3 and infection. All AE were controllable. Conclusion: HDMP+RTX+FFP is an effective and relatively tolerable therapy for patients with B-CLPD accompanying with TP53 abnormalities.
Collapse
Affiliation(s)
- K Shi
- Department of Hematology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing 210029, China
| | | | | | | | | | | | | |
Collapse
|
26
|
PD-1/PD-L1 immune checkpoint and p53 loss facilitate tumor progression in activated B-cell diffuse large B-cell lymphomas. Blood 2019; 133:2401-2412. [PMID: 30975638 DOI: 10.1182/blood.2018889931] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
Refractory or relapsed diffuse large B-cell lymphoma (DLBCL) often associates with the activated B-cell-like (ABC) subtype and genetic alterations that drive constitutive NF-κB activation and impair B-cell terminal differentiation. Here, we show that DNA damage response by p53 is a central mechanism suppressing the pathogenic cooperation of IKK2ca-enforced canonical NF-κB and impaired differentiation resulting from Blimp1 loss in ABC-DLBCL lymphomagenesis. We provide evidences that the interplay between these genetic alterations and the tumor microenvironment select for additional molecular addictions that promote lymphoma progression, including aberrant coexpression of FOXP1 and the B-cell mutagenic enzyme activation-induced deaminase, and immune evasion through major histocompatibility complex class II downregulation, PD-L1 upregulation, and T-cell exhaustion. Consistently, PD-1 blockade cooperated with anti-CD20-mediated B-cell cytotoxicity, promoting extended T-cell reactivation and antitumor specificity that improved long-term overall survival in mice. Our data support a pathogenic cooperation among NF-κB-driven prosurvival, genetic instability, and immune evasion mechanisms in DLBCL and provide preclinical proof of concept for including PD-1/PD-L1 blockade in combinatorial immunotherapy for ABC-DLBCL.
Collapse
|
27
|
Venetoclax plus R- or G-CHOP in non-Hodgkin lymphoma: results from the CAVALLI phase 1b trial. Blood 2019; 133:1964-1976. [PMID: 30850381 DOI: 10.1182/blood-2018-11-880526] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/25/2019] [Indexed: 12/17/2022] Open
Abstract
Novel strategies, such as chemosensitization with targeted agents, that build on the success of standard immunochemotherapy show promise for the treatment of non-Hodgkin lymphoma (NHL). Here, we report a phase 1b study investigating dose escalation of the BCL2 inhibitor, venetoclax, in combination with rituximab or obinutuzumab and cyclophosphamide, doxorubicin, vincristine, and prednisone (R-/G-CHOP) chemotherapy in B-cell NHL. Objectives included safety assessment and determination of a recommended phase 2 dose (RP2D). Fifty-six patients were enrolled, most with follicular lymphoma (43%) or diffuse large B-cell lymphoma (DLBCL; 32%). Dose-limiting toxicities were reported in 3/14 patients at the first venetoclax dose (200 mg/d), after which dosing was changed from daily to 10 days per cycle and escalated to 800 mg. A further reduction to 5 days per cycle occurred at the 800-mg dose level in the G-CHOP arm. Cytopenias were predominant among grade 3/4 events and reported at a higher rate than expected, particularly in the G-CHOP arm; however, safety was manageable. Overall response rates were 87.5% (R-CHOP and G-CHOP combinations); complete response (CR) rates were 79.2% and 78.1%, respectively. Most double-expressor (BCL2+ and MYC+) DLBCL patients (87.5%; n = 7/8) achieved CR. Although the maximum tolerated dose was not reached, the RP2D for venetoclax with R-CHOP was established at 800 mg days 4 to 10 of cycle 1 and days 1 to 10 of cycles 2 to 8; higher doses were not explored, and this dosing schedule demonstrated an acceptable safety profile. This regimen is subsequently being evaluated in first-line DLBCL in the phase 2 portion of the study. This trial was registered at www.clinicaltrials.gov as #NCT02055820.
Collapse
|
28
|
Tessoulin B, Descamps G, Dousset C, Amiot M, Pellat-Deceunynck C. Targeting Oxidative Stress With Auranofin or Prima-1 Met to Circumvent p53 or Bax/Bak Deficiency in Myeloma Cells. Front Oncol 2019; 9:128. [PMID: 30895171 PMCID: PMC6414792 DOI: 10.3389/fonc.2019.00128] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022] Open
Abstract
Prima-1Met (APR-246) was previously shown to be dependent on glutathione inhibition and on ROS induction in cancer cells with mutated or deleted TP53. Because this ROS induction was, at least in part, due to a direct interference with the thioredoxin reductase enzyme, we investigated whether activity of Prima-1Met could be mimicked by auranofin, an inhibitor of the thioredoxin reductase. We thus compared the activity of auranofin and Prima-1Met in 18 myeloma cell lines and in 10 samples from patients with multiple myeloma or plasma cell leukemia. We showed that, similar to Prima-1Met, the activity of auranofin was not dependent on either TP53 status or p53 expression; was inhibited by N-acetyl-L-cysteine, a ROS scavenger; displayed a dramatic synergy with L-buthionine sulfoximine, an irreversible inhibitor of glutathione synthesis; and induced cell death that was not dependent on Bax/Bak expression. These data showed that auranofin and Prima-1Met similarly overcome cell death resistance in myeloma cells due to either p53 deficiency or to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Benoit Tessoulin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,L'Héma-NexT, i-Site NexT, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France.,Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU de Nantes, Nantes, France
| | - Geraldine Descamps
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,L'Héma-NexT, i-Site NexT, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - Christelle Dousset
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,L'Héma-NexT, i-Site NexT, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - Martine Amiot
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,L'Héma-NexT, i-Site NexT, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| | - Catherine Pellat-Deceunynck
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,L'Héma-NexT, i-Site NexT, Université de Nantes, Nantes, France.,SIRIC ILIAD, Angers, Nantes, France
| |
Collapse
|
29
|
Baron M, Simon L, Poulain S, Leblond V. How Recent Advances in Biology of Waldenström's Macroglobulinemia May Affect Therapy Strategy. Curr Oncol Rep 2019; 21:27. [PMID: 30806816 DOI: 10.1007/s11912-019-0768-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Waldenström macroglobulinemia (WM) is a rare lymphoproliferative disorder. Up to now, therapeutic choice was not influenced by the biological characteristics of the disease. Here, we will review how recent advances in biology in WM may affect therapy strategy. RECENT FINDINGS Recently, WM has been described as a new oncogenic model. MyD88 mutation has been described as a key driver mutation and has functional consequences which could be targeted. Other mutations, such as CXCR4 or TP53, have been reported. These mutations are associated with different clinical presentation, prognosis, and treatment response. Mutational status may influence therapeutic choice in some patients but additional data are required. New targeted therapies are on development.
Collapse
Affiliation(s)
- Marine Baron
- Department of Hematology, Pitié-Salpétrière Hospital APHP, Sorbonne Université, Boulevard de l'hôpital, 75013, Paris, France.
| | - Laurence Simon
- Department of Hematology, Centre-Hospitalier Sud-Francilien, Corbeil-Essonnes, France
| | - Stéphanie Poulain
- Department of Cellular Hematology, CHU de Lille, Lille, France.,INSERM UMRX 1172, IRCL, Lille, France
| | - Véronique Leblond
- Department of Hematology, Pitié-Salpétrière Hospital APHP, Sorbonne Université, Boulevard de l'hôpital, 75013, Paris, France
| |
Collapse
|
30
|
High subclonal fraction of 17p deletion is associated with poor prognosis in multiple myeloma. Blood 2019; 133:1217-1221. [PMID: 30692124 DOI: 10.1182/blood-2018-10-880831] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022] Open
Abstract
Deletions of chromosome 17p (del17p) that span the TP53 gene are associated with poor outcome in multiple myeloma (MM), but the prognostic value of del17p cancer clonal fraction (CCF) remains unclear. We applied uniform cytogenetic assessments in a large cohort of newly diagnosed MM (NDMM) patients carrying varying levels of del17p. Incremental CCF change was associated with shorter survival, and a robust CCF threshold of 0.55 was established in discovery and replication data sets. After stratification on the 0.55-CCF threshold, high-risk patients had statistically significantly poorer outcomes compared with low-risk patients (median progression-free survival [PFS] and overall survival [OS], 14 and 32 vs 23.1 and 76.2 months, respectively). Analyses of a third data set comprising whole-exome sequencing data from NDMM patients identified presence of TP53 deletions/mutations as a necessary requirement for high-risk stratification in addition to exceeding the del17p CCF threshold. Meta-analysis conducted across 3 data sets confirmed the robustness of the CCF threshold for PFS and OS. Our analyses demonstrate the feasibility of fluorescence in situ hybridization- and sequencing-based methods to identify TP53 deletions, estimate CCF, and establish that both CCF threshold of 0.55 and presence of TP53 deletion are necessary to identify del17p-carrying NDMM patients with poor prognosis.
Collapse
|
31
|
Tessoulin B, Moreau-Aubry A, Descamps G, Gomez-Bougie P, Maïga S, Gaignard A, Chiron D, Ménoret E, Le Gouill S, Moreau P, Amiot M, Pellat-Deceunynck C. Whole-exon sequencing of human myeloma cell lines shows mutations related to myeloma patients at relapse with major hits in the DNA regulation and repair pathways. J Hematol Oncol 2018; 11:137. [PMID: 30545397 PMCID: PMC6293660 DOI: 10.1186/s13045-018-0679-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/20/2018] [Indexed: 12/28/2022] Open
Abstract
Background Human myeloma cell lines (HMCLs) are widely used for their representation of primary myeloma cells because they cover patient diversity, although not fully. Their genetic background is mostly undiscovered, and no comprehensive study has ever been conducted in order to reveal those details. Methods We performed whole-exon sequencing of 33 HMCLs, which were established over the last 50 years in 12 laboratories. Gene expression profiling and drug testing for the 33 HMCLs are also provided and correlated to exon-sequencing findings. Results Missense mutations were the most frequent hits in genes (92%). HMCLs harbored between 307 and 916 mutations per sample, with TP53 being the most mutated gene (67%). Recurrent bi-allelic losses were found in genes involved in cell cycle regulation (RB1, CDKN2C), the NFκB pathway (TRAF3, BIRC2), and the p53 pathway (TP53, CDKN2A). Frequency of mutations/deletions in HMCLs were either similar to that of patients (e.g., DIS3, PRDM1, KRAS) or highly increased (e.g., TP53, CDKN2C, NRAS, PRKD2). MAPK was the most altered pathway (82% of HMCLs), mainly by RAS mutants. Surprisingly, HMCLs displayed alterations in epigenetic (73%) and Fanconi anemia (54%) and few alterations in apoptotic machinery. We further identified mutually exclusive and associated mutations/deletions in genes involved in the MAPK and p53 pathways as well as in chromatin regulator/modifier genes. Finally, by combining the gene expression profile, gene mutation, gene deletion, and drug response, we demonstrated that several targeted drugs overcome or bypass some mutations. Conclusions With this work, we retrieved genomic alterations of HMCLs, highlighting that they display numerous and unprecedented abnormalities, especially in DNA regulation and repair pathways. Furthermore, we demonstrate that HMCLs are a reliable model for drug screening for refractory patients at diagnosis or at relapse. Electronic supplementary material The online version of this article (10.1186/s13045-018-0679-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Benoît Tessoulin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France. .,Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France.
| | - Agnès Moreau-Aubry
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Géraldine Descamps
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | - Sophie Maïga
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | - David Chiron
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | - Steven Le Gouill
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France
| | - Philippe Moreau
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France.,Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, Nantes, France
| | - Martine Amiot
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | |
Collapse
|
32
|
Lok A, Descamps G, Tessoulin B, Chiron D, Eveillard M, Godon C, Le Bris Y, Vabret A, Bellanger C, Maillet L, Barillé-Nion S, Gregoire M, Fonteneau JF, Le Gouill S, Moreau P, Tangy F, Amiot M, Moreau-Aubry A, Pellat-Deceunynck C. p53 regulates CD46 expression and measles virus infection in myeloma cells. Blood Adv 2018; 2:3492-3505. [PMID: 30530776 PMCID: PMC6290095 DOI: 10.1182/bloodadvances.2018025106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023] Open
Abstract
In this study, we assessed the sensitivity of myeloma cells to the oncolytic measles virus (MV) in relation to p53 using 37 cell lines and 23 primary samples. We showed that infection and cell death were correlated with CD46 expression, which was associated with TP53 status; TP53 abn cell lines highly expressed CD46 and were preferentially infected by MV when compared with the TP53 wt cell lines (P = .046 and P = .045, respectively). Infection of myeloma cells was fully dependent on CD46 expression in both cell lines and primary cells. In the TP53 wt cell lines, but not the TP53 abn cell lines, activation of the p53 pathway with nutlin3a inhibited both CD46 expression and MV infection, while TP53 silencing reciprocally increased CD46 expression and MV infection. We showed using a p53 chromatin immunoprecipitation assay and microRNA assessment that CD46 gene expression was directly and indirectly regulated by p53. Primary myeloma cells overexpressed CD46 as compared with normal cells and were highly infected and killed by MV. CD46 expression and MV infection were inhibited by nutlin3a in primary p53-competent myeloma cells, but not in p53-deficient myeloma cells, and the latter were highly sensitive to MV infection. In summary, myeloma cells were highly sensitive to MV and infection inhibition by the p53 pathway was abrogated in p53-deficient myeloma cells. These results argue for an MV-based clinical trial for patients with p53 deficiency.
Collapse
Affiliation(s)
- Anne Lok
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, and
| | - Geraldine Descamps
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Benoit Tessoulin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, and
| | - David Chiron
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marion Eveillard
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Laboratoire d'Hématologie, CHU de Nantes, Nantes, France
| | | | - Yannick Le Bris
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Laboratoire d'Hématologie, CHU de Nantes, Nantes, France
| | - Astrid Vabret
- National Reference Laboratory for Measles Virus, Département de Virologie, CHU de Caen, Université de Normandie, Caen, France; and
| | - Celine Bellanger
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Laurent Maillet
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Sophie Barillé-Nion
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Marc Gregoire
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | | - Steven Le Gouill
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, and
| | - Philippe Moreau
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, and
| | - Frederic Tangy
- CNRS UMR3569, Unité de Génomique Virale et Vaccination, Institut Pasteur, Paris, France
| | - Martine Amiot
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | - Agnes Moreau-Aubry
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, Nantes, France
| | | |
Collapse
|
33
|
Grewal RK, Chetty M, Abayomi EA, Tomuleasa C, Fromm JR. Use of flow cytometry in the phenotypic diagnosis of hodgkin's lymphoma. CYTOMETRY PART B-CLINICAL CYTOMETRY 2018; 96:116-127. [PMID: 30350336 DOI: 10.1002/cyto.b.21724] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/18/2018] [Accepted: 06/26/2018] [Indexed: 12/22/2022]
Abstract
Hodgkin's lymphoma (HL) has a unique immunophenotype derived from immunohistochemistry (positive for CD15, CD30, and Pax-5; negative for CD3, CD20 in most cases, and CD45). The knowledge gained over recent years enables better diagnosis, prognosis, and treatment of HL. Flow cytometry as a tool for the diagnosis of classic HL has not been useful in the past due to the difficulty in isolating Reed-Sternberg cells as they are admixed in a rich inflammatory background which consists mainly of T cells, B cells, eosinophils, histiocytes, and plasma cells. However, in the recent past, several studies have tried to identify Reed-Sternberg cells using flow cytometry on fine needle aspiration or tissue biopsy of lymph nodes to confirm or supplement immunohistochemistry staining in diagnosis. Newer and more sensitive tools such as flow cytometry can be used for diagnosis, technology that may have been difficult in the past for diagnosis of this lymphoma subtype. Using flow cytometry, diagnosis is faster and could lead to point-of-care technology especially where we have typical immunophenotype signatures. © 2018 International Clinical Cytometry Society.
Collapse
Affiliation(s)
- Ravnit-Kaur Grewal
- MBCHB South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Manogari Chetty
- Department of Oral and Molecular Biology, Faculty of Dentistry, University of the Western Cape, Cape Town, South Africa
| | | | - Ciprian Tomuleasa
- Department of Hematology/Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy-Ion Chiricuta Oncology Institute, Cluj Napoca, Romania
| | - Jonathan R Fromm
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
34
|
Tang C, Kuruvilla J. Optimal frontline management of mantle cell lymphoma: can we agree? Expert Rev Hematol 2018; 11:911-914. [PMID: 30336708 DOI: 10.1080/17474086.2018.1537778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Catherine Tang
- a Division of Medical Oncology & Hematology , Princess Margaret Cancer Centre , Toronto , Canada.,b Department of Medicine , University of Toronto , Toronto , Canada
| | - John Kuruvilla
- a Division of Medical Oncology & Hematology , Princess Margaret Cancer Centre , Toronto , Canada.,b Department of Medicine , University of Toronto , Toronto , Canada
| |
Collapse
|
35
|
Peroja P, Pedersen M, Mantere T, Nørgaard P, Peltonen J, Haapasaari KM, Böhm J, Jantunen E, Turpeenniemi-Hujanen T, Rapakko K, Karihtala P, Soini Y, Vasala K, Kuittinen O. Mutation of TP53, translocation analysis and immunohistochemical expression of MYC, BCL-2 and BCL-6 in patients with DLBCL treated with R-CHOP. Sci Rep 2018; 8:14814. [PMID: 30287880 PMCID: PMC6172218 DOI: 10.1038/s41598-018-33230-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/21/2018] [Indexed: 11/25/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive lymphoma with diverse outcomes. Concurrent translocation of MYC and BCL-2 and/or BCL-6, and concurrent immunohistochemical (IHC) high expression of MYC and BCL-2, have been linked to unfavorable treatment responses. TP53-mutated DLBCL has also been linked to worse outcome. Our aim was to evaluate the aforementioned issues in a cohort of 155 patients uniformly treated with R-CHOP-like therapies. We performed direct sequencing of TP53 exons 5, 6, 7 and 8 as well as fluorescence in-situ hybridization (FISH) of MYC, BCL-2 and BCL-6, and IHC of MYC, BCL-2 and BCL-6. In multivariate analysis, TP53 mutations in L3 and loop-sheet helix (LSH) associated with a risk ratio (RR) of disease-specific survival (DSS) of 8.779 (p = 0.022) and a RR of disease-free survival (DFS) of 10.498 (p = 0.011). In IHC analysis BCL-2 overexpression was associated with inferior DFS (p = 0.002) and DSS (p = 0.002). DLBCL with BCL-2 and MYC overexpression conferred inferior survival in all patients (DSS, p = 0.038 and DFS, p = 0.011) and in patients with non-GC phenotype (DSS (p = 0.013) and DFS (p = 0.010). Our results imply that in DLBCL, the location of TP53 mutations and IHC analysis of BCL-2 and MYC might have a role in the assessment of prognosis.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Murine-Derived
- Antineoplastic Combined Chemotherapy Protocols
- Cyclophosphamide
- Doxorubicin
- Female
- Gene Expression Profiling
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Prednisone
- Proto-Oncogene Proteins c-bcl-2/analysis
- Proto-Oncogene Proteins c-bcl-6/analysis
- Proto-Oncogene Proteins c-myc/analysis
- Rituximab
- Sequence Analysis, DNA
- Survival Analysis
- Translocation, Genetic
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
- Vincristine
- Young Adult
Collapse
Affiliation(s)
- Pekka Peroja
- Department of Oncology and Radiotherapy, Cancer and Translational Medicine Research Unit, University of Oulu and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Mette Pedersen
- Department of Pathology, Herlev and Gentofte University Hospital, University of Copenhagen, Herlev, Denmark
| | - Tuomo Mantere
- Laboratory of Genetics, Northern Finland Laboratory Centre NordLab Oulu, Oulu, Finland
| | - Peter Nørgaard
- Department of Pathology, Herlev and Gentofte University Hospital, University of Copenhagen, Herlev, Denmark
| | - Jenni Peltonen
- Department of Oncology and Radiotherapy, Cancer and Translational Medicine Research Unit, University of Oulu and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | | | - Jan Böhm
- Department of Pathology, Central Finland Central Hospital, Jyväskylä, Finland
| | - Esa Jantunen
- Department of Medicine, University of Eastern Finland/Clinical Medicine, Kuopio University Hospital, Siun Sote -North Carelia Central, Kuopio, Finland
| | - Taina Turpeenniemi-Hujanen
- Department of Oncology and Radiotherapy, Cancer and Translational Medicine Research Unit, University of Oulu and Medical Research Center, Oulu University Hospital, Oulu, Finland
| | - Katrin Rapakko
- Laboratory of Genetics, Northern Finland Laboratory Centre NordLab Oulu, Oulu, Finland
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Cancer and Translational Medicine Research Unit, University of Oulu and Medical Research Center, Oulu University Hospital, Oulu, Finland.
| | - Ylermi Soini
- Department of Pathology, University of Oulu and Medical Research Center, Oulu, Finland
- Department of Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Kaija Vasala
- Department of Oncology and Radiotherapy, Central Finland Central Hospital, Jyväskylä, Finland
| | - Outi Kuittinen
- University of Eastern Finland, Faculty of Health Medicine, Institute of Clinical Medicine Oncology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
36
|
Derenzini E, Rossi A, Treré D. Treating hematological malignancies with drugs inhibiting ribosome biogenesis: when and why. J Hematol Oncol 2018; 11:75. [PMID: 29855342 PMCID: PMC5984324 DOI: 10.1186/s13045-018-0609-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/26/2018] [Indexed: 01/05/2023] Open
Abstract
It is well known that chemotherapy can cure only some cancers in advanced stage, mostly those with an intact p53 pathway. Hematological cancers such as lymphoma and certain forms of leukemia are paradigmatic examples of such scenario. Recent evidence indicates that the efficacy of many of the alkylating and intercalating agents, antimetabolites, topoisomerase, and kinase inhibitors used in cancer therapy is largely due to p53 stabilization and activation consequent to the inhibition of ribosome biogenesis. In this context, innovative drugs specifically hindering ribosome biogenesis showed preclinical activity and are currently in early clinical development in hematological malignancies. The mechanism of p53 stabilization after ribosome biogenesis inhibition is a multistep process, depending on specific factors that can be altered in tumor cells, which can affect the antitumor efficacy of ribosome biogenesis inhibitors (RiBi). In the present review, the basic mechanisms underlying the anticancer activity of RiBi are discussed based on the evidence deriving from available preclinical and clinical studies, with the purpose of defining when and why the treatment with drugs inhibiting ribosomal biogenesis could be highly effective in hematological malignancies.
Collapse
Affiliation(s)
- Enrico Derenzini
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy.
| | - Alessandra Rossi
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy
| | - Davide Treré
- DIMES, Università di Bologna, Via Massarenti 9, Bologna, Italy.
| |
Collapse
|
37
|
Montaño A, Forero-Castro M, Marchena-Mendoza D, Benito R, Hernández-Rivas JM. New Challenges in Targeting Signaling Pathways in Acute Lymphoblastic Leukemia by NGS Approaches: An Update. Cancers (Basel) 2018; 10:cancers10040110. [PMID: 29642462 PMCID: PMC5923365 DOI: 10.3390/cancers10040110] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 12/18/2022] Open
Abstract
The identification and study of genetic alterations involved in various signaling pathways associated with the pathogenesis of acute lymphoblastic leukemia (ALL) and the application of recent next-generation sequencing (NGS) in the identification of these lesions not only broaden our understanding of the involvement of various genetic alterations in the pathogenesis of the disease but also identify new therapeutic targets for future clinical trials. The present review describes the main deletions, amplifications, sequence mutations, epigenetic lesions, and new structural DNA rearrangements detected by NGS in B-ALL and T-ALL and their clinical importance for therapeutic procedures. We reviewed the molecular basis of pathways including transcriptional regulation, lymphoid differentiation and development, TP53 and the cell cycle, RAS signaling, JAK/STAT, NOTCH, PI3K/AKT/mTOR, Wnt/β-catenin signaling, chromatin structure modifiers, and epigenetic regulators. The implementation of NGS strategies has enabled important mutated genes in each pathway, their associations with the genetic subtypes of ALL, and their outcomes, which will be described further. We also discuss classic and new cryptic DNA rearrangements in ALL identified by mRNA-seq strategies. Novel cooperative abnormalities in ALL could be key prognostic and/or predictive biomarkers for selecting the best frontline treatment and for developing therapies after the first relapse or refractory disease.
Collapse
Affiliation(s)
- Adrián Montaño
- IBSAL, IBMCC, Universidad de Salamanca-CSIC, Cancer Research Center, 37007 Salamanca, Spain.
| | - Maribel Forero-Castro
- Escuela de Ciencias Biológicas, Grupo de investigación en Ciencias Biomédicas (GICBUPTC), Universidad Pedagógica y Tecnológica de Colombia, Tunja 150001, Colombia.
| | - Darnel Marchena-Mendoza
- IBSAL, IBMCC, Universidad de Salamanca-CSIC, Cancer Research Center, 37007 Salamanca, Spain.
- Escuela de Ciencias Biológicas, Grupo de investigación en Ciencias Biomédicas (GICBUPTC), Universidad Pedagógica y Tecnológica de Colombia, Tunja 150001, Colombia.
| | - Rocío Benito
- IBSAL, IBMCC, Universidad de Salamanca-CSIC, Cancer Research Center, 37007 Salamanca, Spain.
| | | |
Collapse
|
38
|
Zhong W, Xu X, Zhu Z, Yang L, Du H, Xia Z, Yuan Z, Xiong H, Du Q, Wei Y, Li Q. Increased interleukin-17A levels promote rituximab resistance by suppressing p53 expression and predict an unfavorable prognosis in patients with diffuse large B cell lymphoma. Int J Oncol 2018; 52:1528-1538. [PMID: 29512700 PMCID: PMC5873833 DOI: 10.3892/ijo.2018.4299] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/23/2018] [Indexed: 12/12/2022] Open
Abstract
Rituximab resistance has become increasingly common in patients with diffuse large B cell lymphoma (DLBCL). However, the mechanisms involved remain unclear. In this study, we aimed to examine the effect of rituximab on interleukin (IL)-17A and to investigate the role of IL-17A in rituximab resistance and its prognostic value in patients with DLBCL. Our retrospective analysis revealed that rituximab increased IL-6 expression levels in patients with DLBCL, and the increased IL-6 levels in turn induced the differentiation of Th17 and IL-17+Foxp3+ Treg cells, which secreted IL-17A both in vivo and in vitro. We then examined the effects of IL-17A on the apoptosis and proliferation of, and p53 expression in DLBCL cells, and found that IL-17A prevented rituximab-induced apoptosis and promoted the proliferation of DLBCL cells by suppressing p53 expression in vitro. The survival data of 73 patients with DLBCL suggested that high peripheral blood levels of IL-17A predicted an unfavorable survival. On the whole, our data indicate that rituximab promotes Th17 and IL-17+Foxp3+ Treg cells to secrete IL-17A, which in turn promotes rituximab resistance, partially by suppressing p53 expression and inhibiting rituximab-induced DLBCL cell apoptosis. IL-17A may thus prove to be a useful prognostic marker in patients with DLBCL.
Collapse
Affiliation(s)
- Weijie Zhong
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Xin Xu
- Department of Geriatrics, Hematology and Oncology Ward, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Zhigang Zhu
- Department of Geriatrics, Hematology and Oncology Ward, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Li Yang
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Hong Du
- Department of Pathology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Zhongjun Xia
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Department of Hematological Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Zhaohu Yuan
- Department of Blood Transfusion, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Huabao Xiong
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029-5674, USA
| | - Qinghua Du
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Yaming Wei
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Qingshan Li
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
39
|
Gillardin PS, Descamps G, Maiga S, Tessoulin B, Djamai H, Lucani B, Chiron D, Moreau P, Le Gouill S, Amiot M, Pellat-Deceunynck C, Moreau-Aubry A. Decitabine and Melphalan Fail to Reactivate p73 in p53 Deficient Myeloma Cells. Int J Mol Sci 2017; 19:ijms19010040. [PMID: 29295500 PMCID: PMC5795990 DOI: 10.3390/ijms19010040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/12/2022] Open
Abstract
(1) Background: TP53 deficiency remains a major adverse event in Multiple Myeloma (MM) despite therapeutic progresses. As it is not possible to target TP53 deficiency with pharmacological agents, we explored the possibility of activating another p53 family member, p73, which has not been well studied in myeloma. (2) Methods: Using human myeloma cell lines (HMCLs) with normal or abnormal TP53 status, we assessed TP73 methylation and expression. (3) Results: Using microarray data, we reported that TP73 is weakly expressed in 47 HMCLs and mostly in TP53 wild type (TP53wt) HMCLs (p = 0.0029). Q-RT-PCR assays showed that TP73 was expressed in 57% of TP53wt HMCLs (4 out of 7) and 11% of TP53 abnormal (TP53abn) HMCLs (2 out of 18) (p = 0.0463). We showed that TP73 is silenced by methylation in TP53abn HMCLs and that decitabine increased its expression, which, however, remained insufficient for significant protein expression. Alkylating drugs increased expression of TP73 only in TP53wt HMCLs but failed to synergize with decitabine in TP53abn HMCLs. (4) Conclusions: Decitabine and melphalan does not appear as a promising combination for inducing p73 and bypassing p53 deficiency in myeloma cells.
Collapse
Affiliation(s)
| | - Géraldine Descamps
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | - Sophie Maiga
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | - Benoit Tessoulin
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | - Hanane Djamai
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | - Benedetta Lucani
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | - David Chiron
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | - Philippe Moreau
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, 44093 Nantes, France.
| | - Steven Le Gouill
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
- Service d'Hématologie Clinique, Unité d'Investigation Clinique, CHU, 44093 Nantes, France.
| | - Martine Amiot
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| | | | - Agnès Moreau-Aubry
- CRCINA, INSERM, CNRS, Université d'Angers, Université de Nantes, 44007 Nantes, France.
| |
Collapse
|
40
|
Abdi J, Rastgoo N, Li L, Chen W, Chang H. Role of tumor suppressor p53 and micro-RNA interplay in multiple myeloma pathogenesis. J Hematol Oncol 2017; 10:169. [PMID: 29073933 PMCID: PMC5659022 DOI: 10.1186/s13045-017-0538-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/18/2017] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanisms underlying dysregulated wild type (wt) p53 in multiple myeloma (MM) have been subjects of intense investigation for years. Indeed, correlation of rarely occurring TP53 gene mutations or deletions with adverse clinical outcomes in MM patients is strongly established, while in majority of cases wtp53 seems to be non-functional or dysregulated bearing a high clinical impact. Interestingly, findings from recent investigations show that micro-RNAs (miRNAs) may contribute to suppression of wtp53 in MM, as they are now known to function as key regulatory elements in the p53 network. This area is shedding new light on understanding the biologic effects of dysregulated p53 in MM pathogenesis especially drug resistance. miRNAs such as miR-125b (oncomiR) or miR-34a (tumor suppressor-miR) can be negative or positive regulators of wtp53 function, respectively, with specific effects on MM cell viability. On the other hand, our knowledge of miRNA interaction with mutant (mt) p53 in MM, which is rather related to disease progression and resistance to therapy, is limited which demands in-depth exploration. Here, we will put forward the current knowledge on miRNA-p53 interaction in MM and its role in MM pathogenesis including drug resistance. We will also highlight the pre-clinical approaches for therapeutic application of miRNAs targeting p53 pathway.
Collapse
Affiliation(s)
- Jahangir Abdi
- Division of Molecular and Cellular Biology, Toronto General Research Institute, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | - Nasrin Rastgoo
- Division of Molecular and Cellular Biology, Toronto General Research Institute, Toronto, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada
| | - Lihong Li
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wenming Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hong Chang
- Division of Molecular and Cellular Biology, Toronto General Research Institute, Toronto, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Canada.
- Department of Laboratory Hematology and Medical Oncology, University Health Network, 200 Elizabeth Street, 11E-413, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
41
|
TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 2017; 130:1903-1910. [DOI: 10.1182/blood-2017-04-779736] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022] Open
Abstract
Key Points
The intensified standard-of-care regimens for younger patients with MCL do not overcome the deleterious effects of TP53 mutations. MCLs with TP53 mutations should be considered for alternative frontline treatment.
Collapse
|
42
|
Yoshimoto N, Yanagi T, Matsumura W, Ujiie I, Izumi K, Ando S, Nishie W, Fujii K, Nishihara H, Shimizu H. Primary cutaneous diffuse large B-cell lymphoma presenting as a solitary subcutaneous nodule with TP53 and FBXW7 mutations. Int J Dermatol 2017; 56:1459-1461. [PMID: 28960258 DOI: 10.1111/ijd.13756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/09/2017] [Accepted: 08/16/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Norihiro Yoshimoto
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Teruki Yanagi
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Wakana Matsumura
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Inkin Ujiie
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kentaro Izumi
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satomi Ando
- Department of Dermatology, JCHO Hokushin Hospital, Sapporo, Japan
| | - Wataru Nishie
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kyoko Fujii
- Laboratory of Translational Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroshi Nishihara
- Laboratory of Translational Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroshi Shimizu
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
43
|
Abstract
Abstract
The B-cell leukemia/lymphoma-2 (BCL-2) family of proteins governs the intrinsic pathway of mitochondrial apoptosis. Dysregulation of BCL-2 has long been known to be a crucial part of the pathophysiology of B-cell lymphomas; however, several early attempts to target this pathway therapeutically were unsuccessful because of toxicity, lack of efficacy, or both. Recently, a highly potent and selective oral BCL-2 antagonist, venetoclax, was approved in chronic lymphocytic leukemia, where it has proven to be highly active, even in patients with high-risk del(17p) disease. Venetoclax has also demonstrated efficacy in other B-cell non-Hodgkin lymphoma subtypes, in particular mantle cell lymphoma and follicular lymphoma. Here, I review the history of targeting BCL-2 in B-cell lymphomas, and I discuss recent data on venetoclax used as monotherapy and in combination with monoclonal antibodies, chemotherapy, and other novel agents. I also discuss how genomic and functional approaches such as BH3 profiling may allow us to prioritize novel-agent combinations for further study in clinical trials. These approaches may also help us to understand resistance mechanisms to BCL-2–selective therapy and how to overcome resistance. Finally, I provide my perspective on how to move BCL-2–directed therapies forward toward a goal of developing well-tolerated, time-limited combination regimens with curative potential for patients with B-cell lymphomas.
Collapse
|
44
|
Tisato V, Voltan R, Gonelli A, Secchiero P, Zauli G. MDM2/X inhibitors under clinical evaluation: perspectives for the management of hematological malignancies and pediatric cancer. J Hematol Oncol 2017; 10:133. [PMID: 28673313 PMCID: PMC5496368 DOI: 10.1186/s13045-017-0500-5] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023] Open
Abstract
The two murine double minute (MDM) family members MDM2 and MDMX are at the center of an intense clinical assessment as molecular target for the management of cancer. Indeed, the two proteins act as regulators of P53, a well-known key controller of the cell cycle regulation and cell proliferation that, when altered, plays a direct role on cancer development and progression. Several evidence demonstrated that functional aberrations of P53 in tumors are in most cases the consequence of alterations on the MDM2 and MDMX regulatory proteins, in particular in patients with hematological malignancies where TP53 shows a relatively low frequency of mutation while MDM2 and MDMX are frequently found amplified/overexpressed. The pharmacological targeting of these two P53-regulators in order to restore or increase P53 expression and activity represents therefore a strategy for cancer therapy. From the discovery of the Nutlins in 2004, several compounds have been developed and reported with the ability of targeting the P53-MDM2/X axis by inhibiting MDM2 and/or MDMX. From natural compounds up to small molecules and stapled peptides, these MDM2/X pharmacological inhibitors have been extensively studied, revealing different biological features and different rate of efficacy when tested in in vitro and in vivo experimental tumor models. The data/evidence coming from the preclinical experimentation have allowed the identification of the most promising molecules and the setting of clinical studies for their evaluation as monotherapy or in therapeutic combination with conventional chemotherapy or with innovative therapeutic protocols in different tumor settings. Preliminary results have been recently published reporting data about safety, tolerability, potential side effects, and efficacy of such therapeutic approaches. In this light, the aim of this review is to give an updated overview about the state of the art of the clinical evaluation of MDM2/X inhibitor compounds with a special attention to hematological malignancies and to the potential for the management of pediatric cancers.
Collapse
Affiliation(s)
- Veronica Tisato
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy.
| | - Rebecca Voltan
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Paola Secchiero
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Via Fossato di Mortara 66, 44121, Ferrara, Italy
| |
Collapse
|
45
|
Gong Y, Zhang X, Chen R, Wei Y, Zou Z, Chen X. Cytoplasmic expression of C-MYC protein is associated with risk stratification of mantle cell lymphoma. PeerJ 2017. [PMID: 28626618 PMCID: PMC5472035 DOI: 10.7717/peerj.3457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Aim To investigate the association of C-MYC protein expression and risk stratification in mantle cell lymphoma (MCL), and to evaluate the utility of C-MYC protein as a prognostic biomarker in clinical practice. Methods We conducted immunohistochemical staining of C-MYC, Programmed cell death ligand 1 (PD-L1), CD8, Ki-67, p53 and SRY (sex determining region Y) -11 (SOX11) to investigate their expression in 64 patients with MCL. The staining results and other clinical data were evaluated for their roles in risk stratification of MCL cases using ANOVA, Chi-square, and Spearman’s Rank correlation coefficient analysis. Results Immunohistochemical staining in our study indicated that SOX11, Ki-67 and p53 presented nuclear positivity of tumor cells, CD8 showed membrane positivity in infiltrating T lymphocytes while PD-L1 showed membrane and cytoplasmic positivity mainly in macrophage cells and little in tumor cells. We observed positive staining of C-MYC either in the nucleus or cytoplasm or in both subcellular locations. There were significant differences in cytoplasmic C-MYC expression, Ki-67 proliferative index of tumor cells, and CD8 positive tumor infiltrating lymphocytes (CD8+TIL) among three risk groups (P = 0.000, P = 0.037 and P=0.020, respectively). However, no significant differences existed in the expression of nuclear C-MYC, SOX11, p53, and PD-L1 in MCL patients with low-, intermediate-, and high risks. In addition, patient age and serum LDH level were also significantly different among 3 groups of patients (P = 0.006 and P = 0.000, respectively). Spearman’s rank correlation coefficient analysis indicated that cytoplasmic C-MYC expression, Ki-67 index, age, WBC, as well as LDH level had significantly positive correlations with risk stratification (P = 0.000, 0.015, 0.000, 0.029 and 0.000, respectively), while CD8+TIL in tumor microenvironment negatively correlated with risk stratification of patients (P = 0.006). Patients with increased positive cytoplasmic expression of C-MYC protein and decreased CD8+TIL appeared to be associated with a poor response to chemotherapy, but the correlation was not statistically significant. Conclusion Our study suggested that assessment of cytoplasmic C-MYC overexpression and cytotoxic T lymphocytes (CTLs) by immunohistochemical staining might be helpful for MCL risk stratification and outcome prediction. However, large cohort studies of MCL patients with complete follow up are needed to validate our speculation.
Collapse
Affiliation(s)
- Yi Gong
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China.,Department of Hematology-oncology, Chongqing Cancer Institute/Hospital, Chongqing, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Rui Chen
- Department of Pathology, Chongqing Cancer Institute/Hospital, Chongqing, China
| | - Yan Wei
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongmin Zou
- Institute of Toxicology, School of Preventive Medicine, The Third Military Medical University, Chongqing, China
| | - Xinghua Chen
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|