151
|
Mature lymphoid malignancies: origin, stem cells, and chronicity. Blood Adv 2017; 1:2444-2455. [PMID: 29296894 DOI: 10.1182/bloodadvances.2017008854] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/10/2017] [Indexed: 12/15/2022] Open
Abstract
The chronic behavior of mature lymphoid malignancies, with relapses occurring years apart in many patients, has until recently been unexplained. Patterns of relapse also differ vastly between disease entities, with some being highly curable by chemotherapy whereas others are destined to reemerge after treatment. Lately, the use of next-generation sequencing techniques has revealed essential information on the clonal evolution of lymphoid malignancies. Also, experimental xenograft transplantation point to the possible existence of an ancestral (stem) cell. Such a malignant lymphoid stem cell population could potentially evade current therapies and be the cause of chronicity and death in lymphoma patients; however, the evidence is divergent across disease entities and between studies. In this review we present an overview of genetic studies, case reports, and experimental evidence of the source of mature lymphoid malignancy and discuss the perspectives.
Collapse
|
152
|
Gomes LC, Ferrão ALM, Evangelista FCG, de Almeida TD, Barbosa RC, Carvalho MDG, de Paula Sabino A. Advances in chronic lymphocytic leukemia pharmacotherapy. Biomed Pharmacother 2017; 97:349-358. [PMID: 29091884 DOI: 10.1016/j.biopha.2017.10.105] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/18/2017] [Accepted: 10/21/2017] [Indexed: 12/14/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disease that affects B lymphocytes in most cases. Leukemic lymphocytes have prolonged longevity, defined by resistance to apoptosis. These cells can accumulate in peripheral blood, bone marrow, and solid lymphoid organs. CLL may be indolent or aggressive and has a range of prognostic factors such as expression of CD38 and ZAP-70, immunophenotypic and cytogenetic changes, imbalanced apoptosis proteins, and others. Although CLL has a low mortality rate, this disease is generally not considered curable until today. CLL treatment involves alkylating agents and glucocorticoids, purine analogs, monoclonal antibody therapies, and bone marrow transplantation. In recent decades, new drugs have appeared focusing on new targets and specific molecules, such as the BCR receptor, Bruton's tyrosine kinase, phosphatidylinositol 3-kinase, spleen tyrosine kinase, apoptosis proteins and microRNAs. The most appropriate treatment for CLL is one that involves in its protocol a combination of drugs according to the prognostic factors presented by each patient. In this sense, treatment individualization is essential. This article examines standard treatments for CLL and explores new treatments and potential new targets, as well as schematic protocols to understand where we are, how the treatment has evolved, and the advantages and disadvantages of new targets for CLL therapy.
Collapse
Affiliation(s)
- Lorena Caixeta Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil
| | - Aline Lúcia Menezes Ferrão
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil
| | - Fernanda Cristina Gontijo Evangelista
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil
| | - Tâmara Dauare de Almeida
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil
| | - Rayson Carvalho Barbosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil
| | - Maria das Graças Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil
| | - Adriano de Paula Sabino
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Address: 6627, Presidente Antônio Carlos Ave, Pampulha, Zip Code 31270-901, Belo Horizonte, MG, Brazil.
| |
Collapse
|
153
|
Rosenquist R, Beà S, Du MQ, Nadel B, Pan-Hammarström Q. Genetic landscape and deregulated pathways in B-cell lymphoid malignancies. J Intern Med 2017. [PMID: 28631441 DOI: 10.1111/joim.12633] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With the introduction of next-generation sequencing, the genetic landscape of the complex group of B-cell lymphoid malignancies has rapidly been unravelled in recent years. This has provided important information about recurrent genetic events and identified key pathways deregulated in each lymphoma subtype. In parallel, there has been intense search and development of novel types of targeted therapy that 'hit' central mechanisms in lymphoma pathobiology, such as BTK, PI3K or BCL2 inhibitors. In this review, we will outline the current view of the genetic landscape of selected entities: follicular lymphoma, diffuse large B-cell lymphoma, mantle cell lymphoma, chronic lymphocytic leukaemia and marginal zone lymphoma. We will detail recurrent alterations affecting important signalling pathways, that is the B-cell receptor/NF-κB pathway, NOTCH signalling, JAK-STAT signalling, p53/DNA damage response, apoptosis and cell cycle regulation, as well as other perhaps unexpected cellular processes, such as immune regulation, cell migration, epigenetic regulation and RNA processing. Whilst many of these pathways/processes are commonly altered in different lymphoid tumors, albeit at varying frequencies, others are preferentially targeted in selected B-cell malignancies. Some of these genetic lesions are either involved in disease ontogeny or linked to the evolution of each disease and/or specific clinicobiological features, and some of them have been demonstrated to have prognostic and even predictive impact. Future work is especially needed to understand the therapy-resistant disease, particularly in patients treated with targeted therapy, and to identify novel targets and therapeutic strategies in order to realize true precision medicine in this clinically heterogeneous patient group.
Collapse
Affiliation(s)
- R Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - S Beà
- Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), CIBER de Cáncer, Barcelona, Spain
| | - M-Q Du
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - B Nadel
- CNRS, INSERM, CIML, Aix Marseille University, Marseille, France
| | - Q Pan-Hammarström
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
154
|
Trisomy 12 assessment by conventional fluorescence in-situ hybridization (FISH), FISH in suspension (FISH-IS) and laser scanning cytometry (LSC) in chronic lymphocytic leukemia. Cancer Genet 2017; 216-217:142-149. [PMID: 29025588 DOI: 10.1016/j.cancergen.2017.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/03/2017] [Accepted: 07/26/2017] [Indexed: 01/01/2023]
Abstract
Chronic lymphocytic leukemia (CLL) has an extremely heterogeneous clinical course, and prognostication is based on common genetic abnormalities which are detected by standard cytogenetic methods. However, current methods are restricted by the low number of cells able to be analyzed, resulting in the potential to miss clinically relevant sub-clonal populations of cells. A novel high throughput methodology called fluorescence in situ hybridization in suspension (FISH-IS) incorporates a flow cytometry-based imaging approach with automated analysis of thousands of cells. Here we have demonstrated that the FISH-IS technique is applicable to aneuploidy detection in CLL samples for a range of chromosomes using appropriate centromere probes. This method is able to accurately differentiate between monosomy, disomy and trisomy with a sensitivity of 1% in CLL. An analysis comparing conventional FISH, FISH-IS and laser scanning cytometry (LSC) is presented.
Collapse
|
155
|
Do CH, Bailey S, Macardle C, Thurgood LA, Lower KM, Kuss BJ. Development of locus specific sub-clone separation by fluorescence in situ hybridization in suspension in chronic lymphocytic leukemia. Cytometry A 2017; 91:1088-1095. [PMID: 29024486 DOI: 10.1002/cyto.a.23264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/18/2017] [Accepted: 09/18/2017] [Indexed: 01/02/2023]
Abstract
Intra-tumor genetic heterogeneity is a hallmark of cancer. The ability to monitor and analyze these sub-clonal cell populations can be considered key to successful treatment, particularly in the modern era of targeted therapies. Although advances in sequencing technologies have significantly improved our ability to analyze the mutational landscape of tumors, this utility is reduced when considering small, but clinically significant sub-clones, that is, those representing <10% of the tumor burden. We have developed a high-throughput method that utilizes a 17-probe labeled bacterial artificial chromosome contig to quantify sub-clonal populations of cells based on deletion of a single locus. Chronic lymphocytic leukemia (CLL) cells harboring deletion of the short arm of chromosome 17 (del17p), an important prognostic marker for CLL were used to demonstrate the technique. Sub-clones of del17p cells were quantified and isolated from heterogeneous CLL populations using fluorescence in situ hybridization in suspension (FISH-IS) and the locus specific probe set. Using the combination of FISH-IS with the locus-specific probe set enables automated analysis of tens of thousands of cells, accurately quantifying and isolating cells carrying a del17p. Based on the fluorescence intensity of 17p probes, 17p (TP53) deleted cells were identified and sorted using flow cytometric techniques, and enrichment was demonstrated using single nucleotide polymorphism analysis. The ability to separate sub-clones of cells based on genetic heterogeneity, independent of the clone size, highlights the potential application of this method not only in the diagnostic and prognostic setting, but also as an unbiased approach to enable further detailed genetic analysis of the sub-clone with deep sequencing approaches. © 2017 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Cuc H Do
- Discipline Molecular Medicine and Pathology College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Sheree Bailey
- Department of Immunology Allergy and Arthritis, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Cindy Macardle
- Department of Immunology Allergy and Arthritis, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Lauren A Thurgood
- Discipline Molecular Medicine and Pathology College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Karen M Lower
- Discipline Molecular Medicine and Pathology College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Bryone J Kuss
- Discipline Molecular Medicine and Pathology College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Hematology, Molecular Medicine and Pathology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|
156
|
Monti P, Ghiorzo P, Menichini P, Foggetti G, Queirolo P, Izzotti A, Fronza G. TP63 mutations are frequent in cutaneous melanoma, support UV etiology, but their role in melanomagenesis is unclear. Oncol Rep 2017; 38:1985-1994. [PMID: 28849221 PMCID: PMC5652947 DOI: 10.3892/or.2017.5903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022] Open
Abstract
In contrast to TP53, cancer development is rarely associated with mutations in the TP63 and TP73 genes. Recently, next generation sequencing analysis revealed that TP63 mutations are frequent, specifically in cutaneous melanomas. Cutaneous melanoma represents 4% of skin cancers but it is responsible for 80% of skin cancer related deaths. In the present study, we first determined whether all three members of the P53 family of transcription factors were found mutated in cutaneous melanomas by retrieving all TP53, TP63 and TP73 mutations from cBioPortal (http://www.cbioportal.org/). TP53 and TP63 were frequently mutated [15.0% (91/605) and 14.7% (89/605), respectively], while TP73 [1.5% (9/605)] was more rarely mutated (p<0.0001). A UV-mutation fingerprint was recognized for TP63 and TP73 genes. Then, we tried to evaluate the potential role of TP63 mutations as drivers or passengers in the tumorigenic process. In the former case, the amino acid substitutions should cause significant functional consequences on the main biochemical activity of the P63 protein, namely transactivation. The predicted effects of specific amino acid substitutions by two bioinformatics tools were rather different. Using a yeast-based functional assay, the observed hotspot mutant R379CP63 protein exhibited a substantial residual activity compared to the wild-type (>70%). This result does not support a major role of the mutant P63 protein in melanomagenesis while it is still consistent with the TP63 gene being a recorder of UV exposure. The TP63 mutation spectrum from cutaneous melanomas, when compared with that observed at the germinal level in patients affected by P63-associated diseases [ectodermal dysplasia syndromes, (EDs)], revealed significant differences. The TP63 mutations were more frequent at CpGs sites (p<0.0001) in EDs and at PyPy sites (p<0.0001) in cutaneous melanomas. The two spectra differed significantly (p<0.0001). We conclude that TP63 mutations are frequent in cutaneous melanoma, support UV etiology, but their role in melanomagenesis is unclear.
Collapse
Affiliation(s)
- Paola Monti
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genova, I-16132 Genova, Italy
- Genetics of Rare Cancers Unit, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Paola Menichini
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Giorgia Foggetti
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Paola Queirolo
- Medical Oncology Unit, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| | - Alberto Izzotti
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
- Department of Health Sciences, University of Genova, I-16132 Genova, Italy
| | - Gilberto Fronza
- UOC Mutagenesis, Ospedale Policlinico San Martino, I-16132 Genova, Italy
| |
Collapse
|
157
|
Benedetti D, Tissino E, Pozzo F, Bittolo T, Caldana C, Perini C, Martorelli D, Bravin V, D’Agaro T, Rossi FM, Bomben R, Santinelli E, Zaja F, Pozzato G, Chiarenza A, Di Raimondo F, Del Poeta G, Rossi D, Gaidano G, Dal Bo M, Gattei V, Zucchetto A. NOTCH1 mutations are associated with high CD49d expression in chronic lymphocytic leukemia: link between the NOTCH1 and the NF-κB pathways. Leukemia 2017; 32:654-662. [DOI: 10.1038/leu.2017.296] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/23/2017] [Accepted: 09/08/2017] [Indexed: 12/16/2022]
|
158
|
Giudice ID, Rigolin GM, Raponi S, Cafforio L, Ilari C, Wang J, Bordyuh M, Piciocchi A, Marinelli M, Nanni M, Tavolaro S, Filetti M, Bardi A, Tammiso E, Volta E, Negrini M, Saccenti E, Mauro FR, Rossi D, Gaidano G, Guarini A, Rabadan R, Cuneo A, Foà R. Refined karyotype-based prognostic stratification of chronic lymphocytic leukemia with a low- and very-low-risk genetic profile. Leukemia 2017; 32:543-546. [PMID: 28924243 DOI: 10.1038/leu.2017.292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- I Del Giudice
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - G M Rigolin
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - S Raponi
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - L Cafforio
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - C Ilari
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - J Wang
- Divisions of Life Science and Biomedical Engineering, Hong Kong University of Science and Technology, Hong Kong, China
| | - M Bordyuh
- Department of Biomedical Informatics, Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA
| | - A Piciocchi
- GIMEMA Data Centre, GIMEMA Foundation, Rome, Italy
| | - M Marinelli
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - M Nanni
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - S Tavolaro
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - M Filetti
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - A Bardi
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - E Tammiso
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - E Volta
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - M Negrini
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - E Saccenti
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - F R Mauro
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| | - D Rossi
- Hematology, Oncology Institute of Southern Switzerland and Institute of Oncology Research, Bellinzona, Switzerland
| | - G Gaidano
- Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - A Guarini
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - R Rabadan
- Department of Biomedical Informatics, Center for Computational Biology and Bioinformatics, Columbia University, New York, NY, USA
| | - A Cuneo
- Hematology Section, Azienda Ospedaliera Universitaria Arcispedale S. Anna, University of Ferrara, Ferrara, Italy
| | - R Foà
- Hematology, Department of Cellular Biotechnologies and Hematology, Policlinico Umberto I, Sapienza University, Rome, Italy
| |
Collapse
|
159
|
van den Brand M, Scheijen B, Hess CJ, van Krieken JHJ, Groenen PJTA. Pathways towards indolent B-cell lymphoma - Etiology and therapeutic strategies. Blood Rev 2017; 31:426-435. [PMID: 28802906 DOI: 10.1016/j.blre.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 05/07/2017] [Accepted: 08/04/2017] [Indexed: 02/06/2023]
Abstract
Although patients with indolent B-cell lymphomas have a relatively good survival rate, conventional chemotherapy is not curative. Disease courses are typically characterized by multiple relapses and progressively shorter response duration with subsequent lines of therapy. There has been an explosion of innovative targeted agents in the past years. This review discusses current knowledge on the etiology of indolent B-cell lymphomas with respect to the role of micro-organisms, auto-immune diseases, and deregulated pathways caused by mutations. In particular, knowledge on the mutational landscape of indolent B-cell lymphomas has strongly increased in recent years and harbors great promise for more accurate decision making in the current wide range of therapeutic options. Despite this promise, only in chronic lymphocytic leukemia the detection of TP53 mutations and/or del17p currently have a direct effect on treatment decisions. Nevertheless, it is expected that in the near future the role of genetic testing will increase for prediction of response to targeted treatment as well as for more accurate prediction of prognosis in indolent B-cell lymphomas.
Collapse
MESH Headings
- Animals
- DNA Damage
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/etiology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/microbiology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphoma, B-Cell/etiology
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/microbiology
- Lymphoma, B-Cell/therapy
- Lymphoma, B-Cell, Marginal Zone/etiology
- Lymphoma, B-Cell, Marginal Zone/genetics
- Lymphoma, B-Cell, Marginal Zone/microbiology
- Lymphoma, B-Cell, Marginal Zone/therapy
- Lymphoma, Follicular/etiology
- Lymphoma, Follicular/genetics
- Lymphoma, Follicular/microbiology
- Lymphoma, Follicular/therapy
- Molecular Targeted Therapy/methods
- Mutation
- Signal Transduction
Collapse
Affiliation(s)
- Michiel van den Brand
- Department of Pathology, Radboud university medical center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands; Pathology-DNA, location Rijnstate, Wagnerlaan 55, 6815AD Arnhem, The Netherlands.
| | - Blanca Scheijen
- Department of Pathology, Radboud university medical center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands.
| | - Corine J Hess
- Department of Hematology, Radboud university medical center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands.
| | - J Han Jm van Krieken
- Department of Pathology, Radboud university medical center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands.
| | - Patricia J T A Groenen
- Department of Pathology, Radboud university medical center, Geert Grooteplein Zuid 10, 6525GA Nijmegen, The Netherlands.
| |
Collapse
|
160
|
Poulain S, Roumier C, Bertrand E, Renneville A, Caillault-Venet A, Doye E, Geffroy S, Sebda S, Nibourel O, Nudel M, Herbaux C, Renaud L, Tomowiak C, Guidez S, Tricot S, Roche-Lestienne C, Quesnel B, Preudhomme C, Leleu X. TP53 Mutation and Its Prognostic Significance in Waldenstrom's Macroglobulinemia. Clin Cancer Res 2017; 23:6325-6335. [PMID: 28754818 DOI: 10.1158/1078-0432.ccr-17-0007] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 06/03/2017] [Accepted: 07/18/2017] [Indexed: 11/16/2022]
Abstract
Purpose:TP53 is a tumor-suppressor gene that functions as a regulator influencing cellular responses to DNA damage, and TP53 alterations are associated with pejorative outcome in most B-lymphoid disorders. Little is known regarding TP53 alteration in Waldenstrom's macroglobulinemia (WM).Experimental Design: Here, we have explored the incidence of TP53 alteration using Sanger sequencing and ultradeep-targeted sequencing in 125 WM and 10 immunoglobulin M (IgM) monoclonal gammopathy of undetermined significance (MGUS), along with the clinical features and the associated genomic landscape using single-nucleotide polymorphism array and mutational landscape in an integrative study.Results: Overall, we have identified alteration of TP53 locus including mutation, deletion, and copy-neutral LOH in 11.2% of WM. TP53 mutation was acquired in 7.3% of patients with WM at diagnosis, being absent in IgM MGUS, and was highly correlated to deletion 17p. No correlation with CXCR4 mutations was observed. Patients with TP53 alteration had a greater number of genomic abnormalities. Importantly, WM with TP53 alteration had a significantly shorter overall survival, particularly in symptomatic WM, and independently of the international prognostic scoring system for Waldenstrom macroglobulinemia (IPSSWM) score. Specific treatment for WM with TP53 may have to be studied. Nutlin-3a-targeted p53 signaling induced cytotoxicity preclinically, along with new compounds such as ibrutinib, PrimaMet, or CP31398 that bypass p53 pathway in WM, paving the path for future treatment-tailored options.Conclusions: Our results highlight the clinical significance of detection of TP53 alteration in WM to determine the prognosis of WM and guide the treatment choice. Clin Cancer Res; 23(20); 6325-35. ©2017 AACR.
Collapse
Affiliation(s)
- Stéphanie Poulain
- Service d'Hématologie- Immunologie- Cytogénétique, Centre Hospitalier de Valenciennes, Valenciennes, France.,Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France.,INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Christophe Roumier
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France.,INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Elisabeth Bertrand
- INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Aline Renneville
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France.,INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Aurélie Caillault-Venet
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Emmanuelle Doye
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Sandrine Geffroy
- INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Sheherazade Sebda
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Olivier Nibourel
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France.,INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Morgane Nudel
- Service des Maladies du Sang, Hôpital Huriez, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Charles Herbaux
- Service des Maladies du Sang, Hôpital Huriez, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Loic Renaud
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Cécile Tomowiak
- Service d'Hématologie et Thérapie cellulaire, Centre d'Investigation Clinique, Hôpital de la Milétrie, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,INSERM U1402, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Stéphanie Guidez
- Service d'Hématologie et Thérapie cellulaire, Centre d'Investigation Clinique, Hôpital de la Milétrie, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,INSERM U1402, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Sabine Tricot
- Service d'Hématologie- Immunologie- Cytogénétique, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Catherine Roche-Lestienne
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France.,INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Bruno Quesnel
- INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France.,Service des Maladies du Sang, Hôpital Huriez, Centre Hospitalier Régional et Universitaire de Lille, Lille, France
| | - Claude Preudhomme
- Laboratoire d'Hématologie, Centre de Biologie Pathologie, Centre Hospitalier Régional et Universitaire de Lille, Lille, France.,INSERM UMR 1172, Team 4, Cancer Research Institute of Lille, Lille, France
| | - Xavier Leleu
- Service d'Hématologie et Thérapie cellulaire, Centre d'Investigation Clinique, Hôpital de la Milétrie, Centre Hospitalier Universitaire de Poitiers, Poitiers, France. .,INSERM U1402, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| |
Collapse
|
161
|
Low-burden TP53 mutations in chronic phase of myeloproliferative neoplasms: association with age, hydroxyurea administration, disease type and JAK2 mutational status. Leukemia 2017; 32:450-461. [PMID: 28744014 PMCID: PMC5808067 DOI: 10.1038/leu.2017.230] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 06/30/2017] [Accepted: 07/05/2017] [Indexed: 12/19/2022]
Abstract
The multistep process of TP53 mutation expansion during myeloproliferative neoplasm (MPN) transformation into acute myeloid leukemia (AML) has been documented retrospectively. It is currently unknown how common TP53 mutations with low variant allele frequency (VAF) are, whether they are linked to hydroxyurea (HU) cytoreduction, and what disease progression risk they carry. Using ultra-deep next-generation sequencing, we examined 254 MPN patients treated with HU, interferon alpha-2a or anagrelide and 85 untreated patients. We found TP53 mutations in 50 cases (0.2–16.3% VAF), regardless of disease subtype, driver gene status and cytoreduction. Both therapy and TP53 mutations were strongly associated with older age. Over-time analysis showed that the mutations may be undetectable at diagnosis and slowly increase during disease course. Although three patients with TP53 mutations progressed to TP53-mutated or TP53-wild-type AML, we did not observe a significant age-independent impact on overall survival during the follow-up. Further, we showed that complete p53 inactivation alone led to neither blast transformation nor HU resistance. Altogether, we revealed patient's age as the strongest factor affecting low-burden TP53 mutation incidence in MPN and found no significant age-independent association between TP53 mutations and hydroxyurea. Mutations may persist at low levels for years without an immediate risk of progression.
Collapse
|
162
|
Rodríguez-Vicente AE, Bikos V, Hernández-Sánchez M, Malcikova J, Hernández-Rivas JM, Pospisilova S. Next-generation sequencing in chronic lymphocytic leukemia: recent findings and new horizons. Oncotarget 2017; 8:71234-71248. [PMID: 29050359 PMCID: PMC5642634 DOI: 10.18632/oncotarget.19525] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/12/2017] [Indexed: 11/25/2022] Open
Abstract
The rapid progress in next-generation sequencing technologies has significantly contributed to our knowledge of the genetic events associated with the development, progression and treatment resistance of chronic lymphocytic leukemia patients. Together with the discovery of new driver mutations, next-generation sequencing has revealed an immense degree of both intra- and inter-tumor heterogeneity and enabled us to describe marked clonal evolution. Advances in immunogenetics may be implemented to detect minimal residual disease more sensitively and to track clonal B cell populations, their dynamics and molecular characteristics. The interpretation of these aspects is indispensable to thoroughly examine the genetic background of chronic lymphocytic leukemia. We review and discuss the recent results provided by the different next-generation sequencing techniques used in studying the chronic lymphocytic leukemia genome, as well as future perspectives in the methodologies and applications.
Collapse
Affiliation(s)
- Ana E Rodríguez-Vicente
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom.,IBSAL, IBMCC, Centro de Investigación del Cáncer, Universidad de Salamanca, CSIC, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Vasilis Bikos
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - María Hernández-Sánchez
- IBSAL, IBMCC, Centro de Investigación del Cáncer, Universidad de Salamanca, CSIC, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Jitka Malcikova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine - Hematology and Oncology, Medical Faculty MU and University Hospital, Brno, Czech Republic
| | - Jesús-María Hernández-Rivas
- IBSAL, IBMCC, Centro de Investigación del Cáncer, Universidad de Salamanca, CSIC, Hospital Universitario de Salamanca, Salamanca, Spain.,Hematology Department, Hospital Universitario, Salamanca, Spain.,Department of Medicine, Universidad de Salamanca, Salamanca, Spain
| | - Sarka Pospisilova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic.,Department of Internal Medicine - Hematology and Oncology, Medical Faculty MU and University Hospital, Brno, Czech Republic
| |
Collapse
|
163
|
Rosenquist R, Rosenwald A, Du MQ, Gaidano G, Groenen P, Wotherspoon A, Ghia P, Gaulard P, Campo E, Stamatopoulos K. Clinical impact of recurrently mutated genes on lymphoma diagnostics: state-of-the-art and beyond. Haematologica 2017; 101:1002-9. [PMID: 27582569 DOI: 10.3324/haematol.2015.134510] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/03/2016] [Indexed: 12/14/2022] Open
Abstract
Similar to the inherent clinical heterogeneity of most, if not all, lymphoma entities, the genetic landscape of these tumors is markedly complex in the majority of cases, with a rapidly growing list of recurrently mutated genes discovered in recent years by next-generation sequencing technology. Whilst a few genes have been implied to have diagnostic, prognostic and even predictive impact, most gene mutations still require rigorous validation in larger, preferably prospective patient series, to scrutinize their potential role in lymphoma diagnostics and patient management. In selected entities, a predominantly mutated gene is identified in almost all cases (e.g. Waldenström's macroglobulinemia/lymphoplasmacytic lymphoma and hairy-cell leukemia), while for the vast majority of lymphomas a quite diverse mutation pattern is observed, with a limited number of frequently mutated genes followed by a seemingly endless tail of genes with mutations at a low frequency. Herein, the European Expert Group on NGS-based Diagnostics in Lymphomas (EGNL) summarizes the current status of this ever-evolving field, and, based on the present evidence level, segregates mutations into the following categories: i) immediate impact on treatment decisions, ii) diagnostic impact, iii) prognostic impact, iv) potential clinical impact in the near future, or v) should only be considered for research purposes. In the coming years, coordinated efforts aiming to apply targeted next-generation sequencing in large patient series will be needed in order to elucidate if a particular gene mutation will have an immediate impact on the lymphoma classification, and ultimately aid clinical decision making.
Collapse
Affiliation(s)
- Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Germany and Comprehensive Cancer Center Mainfranken (CCC MF), Germany
| | - Ming-Qing Du
- Division of Molecular Histopathology, Department of Pathology, University of Cambridge, UK
| | - Gianluca Gaidano
- Division of Haematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Patricia Groenen
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Andrew Wotherspoon
- Department of Histopathology, Royal Marsden Hopsital, Fulham Road, London, UK
| | - Paolo Ghia
- Division of Experimental Oncology and Department of Onco-Hematology, Università Vita-Salute San Raffaele and IRCCS Instituto Scientifico San Raffaele, Milan, Italy
| | - Philippe Gaulard
- Department of Pathology, AP-HP, Groupe hospitalier Henri Mondor-Albert Chenevier, Créteil INSERM U955, Université Paris-Est, Créteil, France
| | - Elias Campo
- Hemathopatology Section, Department of Pathology, Hospital Clinic and Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, Spain
| | | | | |
Collapse
|
164
|
Davies NJ, Kwok M, Gould C, Oldreive CE, Mao J, Parry H, Smith E, Agathanggelou A, Pratt G, Taylor AMR, Moss P, Griffiths M, Stankovic T. Dynamic changes in clonal cytogenetic architecture during progression of chronic lymphocytic leukemia in patients and patient-derived murine xenografts. Oncotarget 2017; 8:44749-44760. [PMID: 28496009 PMCID: PMC5546515 DOI: 10.18632/oncotarget.17432] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/29/2017] [Indexed: 01/08/2023] Open
Abstract
Subclonal heterogeneity and clonal selection influences disease progression in chronic lymphocytic leukemia (CLL). It is therefore important that therapeutic decisions are made based on an understanding of the CLL clonal architecture and its dynamics in individual patients. Identification of cytogenetic abnormalities by FISH remains the cornerstone of contemporary clinical practice and provides a simple means for prognostic stratification. Here, we demonstrate that multiplexed-FISH can enhance recognition of CLL subclonal repertoire and its dynamics during disease progression, both in patients and CLL patient-derived xenografts (PDX). We applied a combination of patient-specific FISH probes to 24 CLL cases before treatment and at relapse, and determined putative ancestral relationships between subpopulations with different cytogenetic features. We subsequently established 7 CLL PDX models in NOD/Shi-SCID/IL-2Rγctm1sug/Jic (NOG) mice. Application of multiplexed-FISH to these models demonstrated that all of the identified cytogenetic subpopulations had leukemia propagating activity and that changes in their representation during disease progression could be spontaneous, accelerated by treatment or treatment-induced. We conclude that multiplexed-FISH in combination with PDX models have the potential to distinguish between spontaneous and treatment-induced clonal selection, and therefore provide a valuable tool for the pre-clinical evaluation of novel therapies.
Collapse
MESH Headings
- Animals
- Chromosome Aberrations
- Clonal Evolution/genetics
- Combined Modality Therapy
- Disease Models, Animal
- Disease Progression
- Female
- Heterografts
- Humans
- In Situ Hybridization, Fluorescence
- Karyotyping
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Male
- Mice
- Prognosis
- Single-Cell Analysis
- Treatment Outcome
Collapse
Affiliation(s)
- Nicholas J. Davies
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Marwan Kwok
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Clive Gould
- West Midlands Regional Genetics Laboratory, Birmingham Women's NHS Foundation Trust, Birmingham, UK
| | - Ceri E. Oldreive
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Jingwen Mao
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Helen Parry
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Edward Smith
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Angelo Agathanggelou
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Guy Pratt
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | | | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mike Griffiths
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- West Midlands Regional Genetics Laboratory, Birmingham Women's NHS Foundation Trust, Birmingham, UK
| | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
165
|
Zenz T, Kreuz M, Fuge M, Klapper W, Horn H, Staiger AM, Winter D, Helfrich H, Huellein J, Hansmann ML, Stein H, Feller A, Möller P, Schmitz N, Trümper L, Loeffler M, Siebert R, Rosenwald A, Ott G, Pfreundschuh M, Stilgenbauer S. TP53 mutation and survival in aggressive B cell lymphoma. Int J Cancer 2017; 141:1381-1388. [PMID: 28614910 DOI: 10.1002/ijc.30838] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/08/2017] [Accepted: 04/25/2017] [Indexed: 12/11/2022]
Abstract
TP53 is mutated in 20-25% of aggressive B-cell lymphoma (B-NHL). To date, no studies have addressed the impact of TP53 mutations in prospective clinical trial cohorts. To evaluate the impact of TP53 mutation to current risk models in aggressive B-NHL, we investigated TP53 gene mutations within the RICOVER-60 trial. Of 1,222 elderly patients (aged 61-80 years) enrolled in the study and randomized to six or eight cycles of CHOP-14 with or without Rituximab (NCT00052936), 265 patients were analyzed for TP53 mutations. TP53 mutations were demonstrated in 63 of 265 patients (23.8%). TP53 mutation was associated with higher LDH (65% vs. 37%; p < 0.001), higher international prognostic index-Scores (IPI 4/5 27% vs. 12%; p = 0.025) and B-symptoms (41% vs. 24%; p = 0.011). Patients with TP53 mutation were less likely to obtain a complete remission CR/CRu (CR unconfirmed) 61.9% (mut) vs. 79.7% (wt) (p = 0.007). TP53 mutations were associated with decreased event-free (EFS), progression-free (PFS) and overall survival (OS) (median observation time of 40.2 months): the 3 year EFS, PFS and OS were 42% (vs. 60%; p = 0.012), 42% (vs. 67.5%; p < 0.001) and 50% (vs. 76%; p < 0.001) for the TP53 mutation group. In a Cox proportional hazard analysis adjusting for IPI-factors and treatment arms, TP53 mutation was shown to be an independent predictor of EFS (HR 1.5), PFS (HR 2.0) and OS (HR 2.3; p < 0.001). TP53 mutations are independent predictors of survival in untreated patients with aggressive CD20+ lymphoma. TP53 mutations should be considered for risk models in DLBCL and strategies to improve outcome for patients with mutant TP53 must be developed.
Collapse
Affiliation(s)
- Thorsten Zenz
- Molecular Therapy in Haematology and Oncology and Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (dkfz), Heidelberg, Germany.,Dept. of Medicine V, University Hospital Heidelberg, Heidelberg, Germany.,Dept. of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Markus Kreuz
- Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, Leipzig, Germany
| | - Maxi Fuge
- Dept. of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Wolfram Klapper
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, Universitätsklinikum Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Heike Horn
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, and Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, and University of Tübingen, Stuttgart, Germany
| | - Annette M Staiger
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, and Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, and University of Tübingen, Stuttgart, Germany
| | - Doris Winter
- Dept. of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Hanne Helfrich
- Dept. of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Jennifer Huellein
- Molecular Therapy in Haematology and Oncology and Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Martin-Leo Hansmann
- Dr. Senckenberg Institute of Pathology, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | | | - Alfred Feller
- Institute of Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, and Hämatopathologie Lübeck, Germany
| | - Peter Möller
- Institute of Pathology, Universitätsklinikum Ulm, Ulm, Germany
| | - Norbert Schmitz
- Department of Hematology, Oncology and Pneumonology, University Hospital of Muenster, Muenster, Germany
| | - Lorenz Trümper
- Department of Hematology and Oncology, Georg-August Universität, Göttingen, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Universität Leipzig, Leipzig, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University of Ulm, Ulm, Germany
| | - Andreas Rosenwald
- Institute of Pathology, and Comprehensive Cancer Center Mainfranken (CCC MF), University of Würzburg, Würzburg, Germany
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | | | | | | |
Collapse
|
166
|
Extended follow-up and impact of high-risk prognostic factors from the phase 3 RESONATE study in patients with previously treated CLL/SLL. Leukemia 2017; 32:83-91. [PMID: 28592889 PMCID: PMC5770586 DOI: 10.1038/leu.2017.175] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/11/2017] [Accepted: 05/23/2017] [Indexed: 12/14/2022]
Abstract
In the phase 3 RESONATE study, ibrutinib demonstrated superior progression-free survival (PFS), overall survival (OS) and overall response rate (ORR) compared with ofatumumab in relapsed/refractory CLL patients with high-risk prognostic factors. We report updated results from RESONATE in these traditionally chemotherapy resistant high-risk genomic subgroups at a median follow-up of 19 months. Mutations were detected by Foundation One Heme Panel. Baseline mutations in the ibrutinib arm included TP53 (51%), SF3B1 (31%), NOTCH1 (28%), ATM (19%) and BIRC3 (14%). Median PFS was not reached, with 74% of patients randomized to ibrutinib alive and progression-free at 24 months. The improved efficacy of ibrutinib vs ofatumumab continues in all prognostic subgroups including del17p and del11q. No significant difference within the ibrutinib arm was observed for PFS across most genomic subtypes, although a subset carrying both TP53 mutation and del17p had reduced PFS compared with patients with neither abnormality. Reduced PFS or OS was not evident in patients with only del17p. PFS was significantly better for ibrutinib-treated patients in second-line vs later lines of therapy. The robust clinical activity of ibrutinib continues to show ongoing efficacy and acceptable safety consistent with prior reports, independent of various known high-risk mutations.
Collapse
|
167
|
Ghamlouch H, Nguyen-Khac F, Bernard OA. Chronic lymphocytic leukaemia genomics and the precision medicine era. Br J Haematol 2017; 178:852-870. [DOI: 10.1111/bjh.14719] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hussein Ghamlouch
- Institut National De La Santé Et De La Recherche Médicale (INSERM) U1170; Villejuif France
- Gustave Roussy; Villejuif France
- Université Paris Saclay; Paris France
- Equipe Labellisée Ligue Nationale Contre Le Cancer; Paris France
| | - Florence Nguyen-Khac
- INSERM U1138; Université Pierre et Marie Curie-Paris 6; Service d'Hématologie Biologique; Hôpital Pitié-Salpêtrière; APHP; Paris France
| | - Olivier A. Bernard
- Institut National De La Santé Et De La Recherche Médicale (INSERM) U1170; Villejuif France
- Gustave Roussy; Villejuif France
- Université Paris Saclay; Paris France
- Equipe Labellisée Ligue Nationale Contre Le Cancer; Paris France
| |
Collapse
|
168
|
Diffuse large B-cell lymphoma genotyping on the liquid biopsy. Blood 2017; 129:1947-1957. [DOI: 10.1182/blood-2016-05-719641] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 01/11/2017] [Indexed: 12/27/2022] Open
Abstract
Key Points
Plasma cfDNA genotyping is as accurate as genotyping of the diagnostic biopsy in detecting clonal somatic mutations in DLBCL. Plasma cfDNA genotyping is a real-time, noninvasive tool that can be used to track clonal evolution in DLBCL.
Collapse
|
169
|
Huang SJ, Bergin K, Smith AC, Gerrie AS, Bruyere H, Dalal CB, Sugioka DK, Hrynchak M, Ramadan KM, Karsan A, Gillan TL, Toze CL. Clonal evolution as detected by interphase fluorescence in situ hybridization is associated with worse overall survival in a population-based analysis of patients with chronic lymphocytic leukemia in British Columbia, Canada. Cancer Genet 2017; 210:1-8. [PMID: 28212806 DOI: 10.1016/j.cancergen.2016.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/20/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022]
Abstract
This study evaluates prognostic markers as predictors of clonal evolution (CE) and assesses the impact of CE on overall survival (OS) in a population-based cohort of 159 consecutive eligible patients with chronic lymphocytic leukemia (CLL) obtained from the British Columbia Provincial CLL Database. CE was detected by interphase fluorescence in situ hybridization (FISH) in 34/159 patients (21%) with 65% of CE patients acquiring deletion 17p or 11q. CD38 positive status (≥30%) on flow cytometry predicted 2.7 times increased risk of high-risk CE (acquisition of deletion 17p or 11q) on multivariate analysis. Prior CLL therapy was not a significant predictor of CE. CE was associated with 4.1 times greater risk of death when analyzed as a time-dependent variable for OS after adjusting for age, lymphocyte count, and FISH timing. High-risk CE was associated with worse OS while acquisition of low/intermediate-risk abnormalities (trisomy 12, deletion 13q, and IGH translocation) had no difference in OS. Our study demonstrates the negative impact of CE detected by FISH on OS in this population-based cohort. These data provide support for repeating FISH testing during CLL follow-up as patients with high-risk CE have reduced survival and may require closer observation.
Collapse
Affiliation(s)
- Steven J Huang
- Division of Hematology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada; Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Krystal Bergin
- Division of Hematology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Adam C Smith
- Cancer Genetics Laboratory, Pathology and Laboratory Medicine, British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada; The Pelé Research Institute, Hospital Pequeno Prinicipe, Brazil
| | - Alina S Gerrie
- Division of Hematology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada; Leukemia/BMT Program of BC, Vancouver General Hospital and British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - Helene Bruyere
- Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Chinmay B Dalal
- Division of Hematology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | | | - Monica Hrynchak
- Cytogenetics Laboratory, Royal Columbian Hospital, New Westminster, BC, Canada
| | - Khaled M Ramadan
- Division of Hematology, St. Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Aly Karsan
- Cancer Genetics Laboratory, Pathology and Laboratory Medicine, British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada
| | - Tanya L Gillan
- Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Cynthia L Toze
- Division of Hematology, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada; Leukemia/BMT Program of BC, Vancouver General Hospital and British Columbia Cancer Agency, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
170
|
Lazarian G, Guièze R, Wu CJ. Clinical Implications of Novel Genomic Discoveries in Chronic Lymphocytic Leukemia. J Clin Oncol 2017; 35:984-993. [PMID: 28297623 DOI: 10.1200/jco.2016.71.0822] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a common B-cell malignancy with a remarkably heterogeneous course, ranging from indolent disease with no need for immediate therapy to rapidly progressive disease associated with therapeutic resistance. The recent US Food and Drug Administration approvals of novel targeted therapies such as inhibitors of B-cell receptor signaling and B-cell lymphoma 2 have opened up new opportunities in the clinical management of patients with CLL and heralded a new era in the clinical treatment of this disease. In parallel, the implementation of novel sequencing technologies has provided new insights into CLL complexity, identifying a growing list of putative drivers that underlie inter- and intratumor heterogeneities in CLL affecting disease progression and resistance. The identification of these novel genomic features that can indicate future drug resistance or guide therapeutic management is now becoming a major goal in CLL so that patients can best benefit from the increasingly diverse available therapies, as discussed herein.
Collapse
Affiliation(s)
- Gregory Lazarian
- All authors: Dana-Farber Cancer Institute; Romain Guièze and Catherine J. Wu, Harvard Medical School; Catherine J. Wu, Brigham and Women's Hospital, Boston; Romain Guièze and Catherine J. Wu, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; Gregory Lazarian, U978 Institut National de la Santé et de la Recherche Médicale and Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Bobigny; and Romain Guièze, Centre Hospitalier Universitaire de Clermont-Ferrand and Université d'Auvergne, Clermont-Ferrand, France
| | - Romain Guièze
- All authors: Dana-Farber Cancer Institute; Romain Guièze and Catherine J. Wu, Harvard Medical School; Catherine J. Wu, Brigham and Women's Hospital, Boston; Romain Guièze and Catherine J. Wu, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; Gregory Lazarian, U978 Institut National de la Santé et de la Recherche Médicale and Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Bobigny; and Romain Guièze, Centre Hospitalier Universitaire de Clermont-Ferrand and Université d'Auvergne, Clermont-Ferrand, France
| | - Catherine J Wu
- All authors: Dana-Farber Cancer Institute; Romain Guièze and Catherine J. Wu, Harvard Medical School; Catherine J. Wu, Brigham and Women's Hospital, Boston; Romain Guièze and Catherine J. Wu, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA; Gregory Lazarian, U978 Institut National de la Santé et de la Recherche Médicale and Hôpital Avicenne, Assistance Publique-Hôpitaux de Paris, Bobigny; and Romain Guièze, Centre Hospitalier Universitaire de Clermont-Ferrand and Université d'Auvergne, Clermont-Ferrand, France
| |
Collapse
|
171
|
Amir H, Touboul T, Sabatini K, Chhabra D, Garitaonandia I, Loring JF, Morey R, Laurent LC. Spontaneous Single-Copy Loss of TP53 in Human Embryonic Stem Cells Markedly Increases Cell Proliferation and Survival. Stem Cells 2017; 35:872-885. [PMID: 27888558 DOI: 10.1002/stem.2550] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 10/17/2016] [Accepted: 11/01/2016] [Indexed: 01/17/2023]
Abstract
Genomic aberrations have been identified in many human pluripotent stem cell (hPSC) cultures. Commonly observed duplications in portions of chromosomes 12p and 17q have been associated with increases in genetic instability and resistance to apoptosis, respectively. However, the phenotypic consequences related to sporadic mutations have not been evaluated to date. Here, we report on the effects of a single-copy deletion of the chr17p13.1 region, a sporadic mutation that spontaneously arose independently in several subclones of a human embryonic stem cell culture. Compared to cells with two normal copies of chr17p13.1 ("wild-type"), the cells with a single-copy deletion of this region ("mutant") displayed a selective advantage when exposed to stressful conditions, and retained a higher percentage of cells expressing the pluripotency marker POU5F1/OCT4 after 2 weeks of in vitro differentiation. Knockdown of TP53, which is a gene encompassed by the deleted region, in wild-type cells mimicked the chr17p13.1 deletion phenotype. Thus, sporadic mutations in hPSCs can have phenotypic effects that may impact their utility for clinical applications. Stem Cells 2017;35:872-885.
Collapse
Affiliation(s)
- Hadar Amir
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| | - Thomas Touboul
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| | - Karen Sabatini
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| | - Divya Chhabra
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| | - Ibon Garitaonandia
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Jeanne F Loring
- Center for Regenerative Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | - Robert Morey
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| | - Louise C Laurent
- Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
172
|
De Paoli L, Gaidano G. Chronic lymphocytic leukaemia: a step ahead in the journey toward eradication. Lancet Oncol 2017; 18:163-164. [PMID: 28089631 DOI: 10.1016/s1470-2045(17)30017-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/04/2017] [Indexed: 11/29/2022]
Affiliation(s)
- Lorenzo De Paoli
- Division of Haematology, Department of Translational Medicine, University of Eastern Piedmont and Maggiore Charity Hospital, 28100 Novara, Italy
| | - Gianluca Gaidano
- Division of Haematology, Department of Translational Medicine, University of Eastern Piedmont and Maggiore Charity Hospital, 28100 Novara, Italy.
| |
Collapse
|
173
|
Pospisilova S, Sutton LA, Malcikova J, Tausch E, Rossi D, Montserrat E, Moreno C, Stamatopoulos K, Gaidano G, Rosenquist R, Ghia P. Innovation in the prognostication of chronic lymphocytic leukemia: how far beyond TP53 gene analysis can we go? Haematologica 2017; 101:263-5. [PMID: 26928246 DOI: 10.3324/haematol.2015.139246] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Sarka Pospisilova
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lesley-Ann Sutton
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Jitka Malcikova
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Eugen Tausch
- Department of Internal Medicine III, Ulm University, Germany
| | - Davide Rossi
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Emili Montserrat
- Institute of Hematology and Oncology, Department of Hematology, Hospital Clinic, University of Barcelona, Spain
| | - Carol Moreno
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Kostas Stamatopoulos
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden Institute of Applied Biosciences, CERTH, Thessaloniki, Greece
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Paolo Ghia
- Division of Experimental Oncology and Department of Onco-Hematology, Università Vita-Salute San Raffaele and IRCCS Instituto Scientifico San Raffaele, Milan, Italy
| | | |
Collapse
|
174
|
Li H, Xiong W, Liu H, Yi S, Yu Z, Liu W, Lyu R, Wang T, Zou D, Li Z, Qiu L. Serum LDH level may predict outcome of chronic lymphocytic leukemia patients with a 17p deletion: a retrospective analysis of prognostic factors in China. Chin J Cancer Res 2017; 29:156-165. [PMID: 28536495 PMCID: PMC5422418 DOI: 10.21147/j.issn.1000-9604.2017.02.09] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective This study aims to evaluate the natural history of patients with chronic lymphocytic leukemia (CLL) and a 17p deletion (17p-) and identify the predictive factors within this subgroup. Methods The sample of patients with CLL were analyzed by fluorescencein situ hybridization for deletions in chromosome bands 11q22, 13q14 and 17p13; trisomy of bands 12q13; and translocation involving band 14q32. The data from 456 patients with or without a 17p- were retrospectively collected and analyzed.
Results The overall response rate (ORR) in patients with a 17p- was 56.9%, and patients with a high percentage of 17p- (defined as more than 25% of cells harbouring a 17p-) had a lower ORR. The median overall survival (OS) in patients with a 17p- was 78.0 months, which was significantly shorter than the OS in patients without this genetic abnormality (median 162.0 months, P<0.001). Within the subgroup with a 17p-, the progression-free survival was significantly shorter in patients at Binet stage B-C and patients with elevated lactate dehydrogenase (LDH), B symptoms, unmutatedIGHV and a high percentage of 17p-.
Conclusions These results indicated that patients with a 17p- CLL have a variable prognosis that might be predicted using simple clinical and laboratory characteristics.
Collapse
Affiliation(s)
- Heng Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Wenjie Xiong
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Huimin Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Rui Lyu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Tingyu Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Zengjun Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
175
|
Abstract
The recent application of next-generation sequencing technologies lead to significant improvements in our understanding of genetic underpinnings of non-Hodgkin lymphomas with identification of an unexpectedly high number of novel mutation targets across the different B-cell lymphoma entities. These recently discovered molecular lesions are expected to have a major impact on development of novel biomarkers and targeted therapies as well as patient stratification based on the underlying genetic profile. This review will cover the major discoveries in B-cell lymphomas using next-generation sequencing technologies over the last few years, highlighting alterations associated with relapse and progression of these diseases.
Collapse
Affiliation(s)
- Csaba Bödör
- MTA-SE Lendulet Molecular Oncohematology Research Group, Budapest, Hungary.,1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Lilla Reiniger
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary. .,2nd Department of Pathology, MTA-SE NAP, Brain Metastasis Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
176
|
Minimal residual disease is an independent predictor for 10-year survival in CLL. Blood 2016; 128:2770-2773. [DOI: 10.1182/blood-2016-05-714162] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 09/26/2016] [Indexed: 11/20/2022] Open
Abstract
Key Points
MRD negativity is a predictor for long-term progression-free and overall survival independent of the type and line of therapy. MRD negativity confers the greatest prognostic benefit when achieved in the frontline setting.
Collapse
|
177
|
Abstract
The term Richter syndrome (RS) indicates the transformation of chronic lymphocytic leukemia (CLL) into an aggressive lymphoma. RS is a rare complication with an aggressive clinical course, bearing an unfavorable prognosis. In the majority of cases, CLL transforms into RS as diffuse large B cell lymphoma (DLBCL), and a clonal relation between the two processes can be found. However, clonally unrelated RS can occur and transformations to other histologies beside DLBCL have been described. Recent data have shed some light on genetic characteristics that can influence and drive the transformation from CLL to RS. This molecular information has not been translated yet into significant treatment advances, and currently the therapy regimens for RS continue to rely on intensive chemotherapy combinations followed by stem cell transplant in suitable candidates. Based on the rapid pace of discoveries in the field of hematological malignancies and on the recent revolution in the therapeutic landscape for CLL and B cell lymphomas, new therapeutic options for RS might be available in the upcoming years.
Collapse
Affiliation(s)
- Candida Vitale
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Unit 428, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Unit 428, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
178
|
Eichhorst B, Hallek M. Prognostication of chronic lymphocytic leukemia in the era of new agents. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2016; 2016:149-155. [PMID: 27913474 PMCID: PMC6142472 DOI: 10.1182/asheducation-2016.1.149] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The prognosis of chronic lymphocytic leukemia (CLL) is very heterogeneous. Therefore, a plethora of prognostic factors has been identified to allow a better prediction of the individual prognosis of a given patient. The clinical staging systems by Rai and Binet have been the backbone of clinical management for several decades. The advent of genetic and biochemical markers, as well as next-generation sequencing has provided several markers that can predict the prognosis of patients with CLL. Using this knowledge, several scores have been created to improve predicting overall survival and/or treatment-free survival. These prognostic scores were developed in the era of chemotherpay/chemoimmunotherapy. Therefore, they now need to be tested with novel agents. However, despite tremendously improved therapeutic options, CLL patients with TP53 dysfunction or a complex karyotype remain at very high risk and seem to have a shorter (treatment-free) survival. The recently published international prognostic index (CLL IPI) incorporates most of these factors and provides a tool to analyze outcome in the modern era of targeted therapies.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Disease-Free Survival
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Survival Rate
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Barbara Eichhorst
- Department I for Internal Medicine and Centre of Integrated Oncology, and
| | - Michael Hallek
- Department I for Internal Medicine and Centre of Integrated Oncology, and
- CECAD—Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
179
|
Identifying High-Risk Chronic Lymphocytic Leukemia: A Pathogenesis-Oriented Appraisal of Prognostic and Predictive Factors in Patients Treated with Chemotherapy with or without Immunotherapy. Mediterr J Hematol Infect Dis 2016; 8:e2016047. [PMID: 27872727 PMCID: PMC5111525 DOI: 10.4084/mjhid.2016.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 09/16/2016] [Indexed: 11/21/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) displays an extremely variable clinical behaviour. Accurate prognostication and prediction of response to treatment are important in an era of effective first-line regimens and novel molecules for high risk patients. Because a plethora of prognostic biomarkers were identified, but few of them were validated by multivariable analysis in comprehensive prospective studies, we applied in this survey stringent criteria to select papers from the literature in order to identify the most reproducible prognostic/predictive markers. Each biomarker was analysed in terms of reproducibility across the different studies with respect to its impact on time to first treatment (TTFT), progression free survival (PFS), overall survival (OS) and response to treatment. We were able to identify the following biomarkers as the most reliable in guiding risk stratification in the daily clinical practice: 17p-/TP53 mutations, IGHV unmutated configuration, short telomeres and 11q-. However, the method for measuring telomere length was not validated yet and 11q- was predictive of inferior OS only in those patients who did not receive FCR-like combinations. Stage and lymphocytosis were predictive of shorter TTFT and age, high serum thymidine kinase levels and poor performance status were predictive of shorter OS. Using our criteria no parameter was found to independently predict for inferior response to treatment.
Collapse
|
180
|
Wooler G, Melchior L, Ralfkiaer E, Rahbek Gjerdrum LM, Gniadecki R. TP53 Gene Status Affects Survival in Advanced Mycosis Fungoides. Front Med (Lausanne) 2016; 3:51. [PMID: 27891503 PMCID: PMC5104736 DOI: 10.3389/fmed.2016.00051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 10/19/2016] [Indexed: 12/27/2022] Open
Abstract
TP53 is frequently mutated in different types of neoplasms including leukemia and lymphomas. Mutations of TP53 have also been reported in mycosis fungoides (MF), the most common type of cutaneous lymphoma. However, little is known about the frequency, spectrum of mutations, and their prognostic significance in MF. In this study, we have optimized the protocol for Sanger sequencing of TP53 using DNA extracted from archival paraffin-embedded biopsies. Of 19 samples from patients with stage IIB MF or higher, 31% harbored mutations in TP53. Overall survival of the patients with mutated TP53 was significantly shorter than median survival in the age- and stage-matched patients treated in our Institution. Distribution of mutations was heterogenous in TP53 exons; however, C > T transitions were common suggesting the causal role of ultraviolet radiation. We propose that TP53 mutation status would be useful for risk stratification of patients with advanced MF.
Collapse
Affiliation(s)
- Gitte Wooler
- Department of Pathology, Zealand University Hospital , Roskilde , Denmark
| | - Linea Melchior
- Department of Pathology, Rigshospitalet , Copenhagen , Denmark
| | | | | | - Robert Gniadecki
- Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark; Division of Dermatology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
181
|
Rossi D, Gerber B, Stüssi G. Predictive and prognostic biomarkers in the era of new targeted therapies for chronic lymphocytic leukemia. Leuk Lymphoma 2016; 58:1548-1560. [PMID: 27808579 DOI: 10.1080/10428194.2016.1250264] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Treatment options for chronic lymphocytic leukemia (CLL) have improved with the introduction of the B-cell receptor inhibitors ibrutinib and idelalisib, and of the BCL2 inhibitor venetoclax. While awaiting the results of head to head comparisons between novel agents and chemoimmunotherapy, predictive biomarkers can assist physicians in treatment tailoring. Though novel agents have modified the landscape of predictors at the time of treatment requirement, the usefulness of historical CLL prognostic biomarkers is still up-to-date when considering anticipation of time to first treatment. This review discusses: (i) disease-related (TP53 defects, immunoglobulin gene mutations), therapy-related (duration of remission), and patient-related (age, comorbidities) biomarkers that can be used in the clinical practice to inform CLL treatment decision either at the time of first line therapy and disease relapse; and (ii) the need of new biomarkers to re-define high-risk CLL because of the questioning by novel agents of historical prognostic factors.
Collapse
Affiliation(s)
- Davide Rossi
- a Hematology , Oncology Institute of Southern Switzerland , Bellinzona , Switzerland.,b Institute of Oncology Research , Bellinzona , Switzerland
| | - Bernhard Gerber
- a Hematology , Oncology Institute of Southern Switzerland , Bellinzona , Switzerland
| | - Georg Stüssi
- a Hematology , Oncology Institute of Southern Switzerland , Bellinzona , Switzerland
| |
Collapse
|
182
|
The mutational signature of chronic lymphocytic leukemia. Biochem J 2016; 473:3725-3740. [DOI: 10.1042/bcj20160256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 08/23/2016] [Indexed: 01/14/2023]
Abstract
Advances in next-generation sequencing technologies continue to unravel the cancer genome, identifying key biological pathways important for disease pathogenesis and clinically relevant genetic lesions. These studies have provided unprecedented resolution of the cancer genome, facilitating significant advances in the ability to detect many cancers, and predict patients who will develop an aggressive disease or respond poorly to treatment. The mature B-cell neoplasm chronic lymphocytic leukaemia remains at the forefront of these genomic analyses, largely due its protracted natural history and the accessibility to suitable material for study. We now possess a comprehensive view of the genomic copy number mutational landscape of the disease, as well as a detail description of clonal evolution, and the molecular mechanisms that drive the acquisition of genomic lesions and more broadly, genomic complexity. Here, recent genomic insights with associated biological and clinical implications will be reviewed.
Collapse
|
183
|
Genetic evolution in chronic lymphocytic leukaemia. Best Pract Res Clin Haematol 2016; 29:67-78. [PMID: 27742073 DOI: 10.1016/j.beha.2016.08.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 06/30/2016] [Accepted: 08/04/2016] [Indexed: 11/21/2022]
Abstract
Next-generation sequencing provides a comprehensive understanding of the genomic, epigenomic and transcriptomic underpinnings of chronic lymphocytic leukaemia. Recent studies have uncovered new drivers, including mutations in non-coding regions, and signalling pathways whose role in cancer was previously unknown or poorly understood. Moreover, massive scale epigenomics and transcriptomics have supplied the foundations for the cellular origin of the disease. Some drivers could be targeted pharmacologically, and the ability to detect mutations present in minority subclones might even allow treatment before clonal selection occurs, thus preventing disease refractoriness. As our understanding broadens and ongoing technological innovation propels new achievements, we will certainly learn how to apply it in our daily practice.
Collapse
|
184
|
Ladetto M, Buske C, Hutchings M, Dreyling M, Gaidano G, Le Gouill S, Luminari S, Pott C, Zamò A, Zucca E. ESMO consensus conference on malignant lymphoma: general perspectives and recommendations for prognostic tools in mature B-cell lymphomas and chronic lymphocytic leukaemia. Ann Oncol 2016; 27:2149-2160. [PMID: 27701070 DOI: 10.1093/annonc/mdw419] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/11/2016] [Accepted: 08/23/2016] [Indexed: 01/02/2023] Open
Abstract
The European Society for Medical Oncology (ESMO) consensus conference on mature B-cell lymphomas and chronic lymphocytic leukaemia (CLL) was held on 20 June 2015 in Lugano, Switzerland, and included a multidisciplinary panel of 25 leading experts. The aim of the conference was to develop recommendations on critical subjects difficult to consider in detail in the ESMO Clinical Practice Guidelines. The following areas were identified: (i) the elderly patient, (ii) prognostic factors suitable for clinical use and (iii) the 'ultra-high-risk' group. Before the conference, the expert panel was divided into three working groups; each group focused on one of these areas in order to address four clinically relevant questions relating to that topic. All relevant scientific literature, as identified by the experts, was reviewed in advance. During the consensus conference, each working group developed recommendations to address each of the four questions assigned to their group. These recommendations were then presented to the entire panel and a consensus was reached. This manuscript presents recommendations dedicated to the second area of interest, i.e. prognostic factors suitable for clinical use. The four topics [i.e. interim positron emission tomography (PET), TP53 mutations, cell of origin (COO) and minimal residual disease (MRD)] were primarily chosen because of the bulk of available data together with the lack of clear guidance regarding their use in clinical practice and within clinical trials. Results, including a summary of evidence supporting each recommendation, are detailed in this manuscript. The panel acknowledged that detection of TP53 inactivation by deletion or mutation in CLL should be implemented in clinical practice (level of evidence I, strength of recommendation A). Due to their potentially high prognostic value, at least in some lymphoma entities, implementation of interim PET, COO and MRD was highly recommended in the context of clinical trials. All expert panel members approved this final article.
Collapse
Affiliation(s)
- M Ladetto
- Hematology Division, Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - C Buske
- Comprehensive Cancer Center Ulm and Department of Internal Medicine III, Institute of Experimental Cancer Research, University Hospital, Ulm, Germany
| | - M Hutchings
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - M Dreyling
- Medizinische Klinik III, Klinikum der Universität München/LMU, Munich, Germany
| | - G Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - S Le Gouill
- Clinical Hematology, Centre Hospitalo-Universitaire de Nantes, UMR892 Team 10, CIC Nantes, France
| | - S Luminari
- Hematology, Arcispedale S. Maria Nuova, IRCCS Reggio Emilia.,Department of Diagnostic, Clinical and Public Health Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - C Pott
- Second Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - A Zamò
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - E Zucca
- Lymphoma Unit, Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | | |
Collapse
|
185
|
Deciphering KRAS and NRAS mutated clone dynamics in MLL-AF4 paediatric leukaemia by ultra deep sequencing analysis. Sci Rep 2016; 6:34449. [PMID: 27698462 PMCID: PMC5048141 DOI: 10.1038/srep34449] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/09/2016] [Indexed: 12/28/2022] Open
Abstract
To induce and sustain the leukaemogenic process, MLL-AF4+ leukaemia seems to require very few genetic alterations in addition to the fusion gene itself. Studies of infant and paediatric patients with MLL-AF4+ B cell precursor acute lymphoblastic leukaemia (BCP-ALL) have reported mutations in KRAS and NRAS with incidences ranging from 25 to 50%. Whereas previous studies employed Sanger sequencing, here we used next generation amplicon deep sequencing for in depth evaluation of RAS mutations in 36 paediatric patients at diagnosis of MLL-AF4+ leukaemia. RAS mutations including those in small sub-clones were detected in 63.9% of patients. Furthermore, the mutational analysis of 17 paired samples at diagnosis and relapse revealed complex RAS clone dynamics and showed that the mutated clones present at relapse were almost all originated from clones that were already detectable at diagnosis and survived to the initial therapy. Finally, we showed that mutated patients were indeed characterized by a RAS related signature at both transcriptional and protein levels and that the targeting of the RAS pathway could be of beneficial for treatment of MLL-AF4+ BCP-ALL clones carrying somatic RAS mutations.
Collapse
|
186
|
Dubuc AM, Davids MS, Pulluqi M, Pulluqi O, Hoang K, Hernandez-Sánchez JM, Schlich C, Hernández-Rivas JM, Brown JR, Dal Cin P. FISHing in the dark: How the combination of FISH and conventional karyotyping improves the diagnostic yield in CpG-stimulated chronic lymphocytic leukemia. Am J Hematol 2016; 91:978-83. [PMID: 27341486 DOI: 10.1002/ajh.24452] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/26/2016] [Accepted: 06/17/2016] [Indexed: 01/01/2023]
Abstract
Despite significant advances in molecular genetic approaches, fluorescence in situ hybridization (FISH) remains the gold standard for the diagnostic evaluation of genomic aberrations in patients with chronic lymphocytic leukemia (CLL). Efforts to improve the diagnostic utility of molecular cytogenetic testing have led to the expansion of the traditional 4-probe CLL FISH panel. Not only do these efforts increase the cost of testing, they remain hindered by the inherent limitations of FISH studies - namely the inability to evaluate genomic changes outside of the targeted loci. While array-based profiling and next generation sequencing (NGS) have critically expanded our understanding of the molecular pathogenesis of CLL, these methodologies are not routinely used by diagnostic laboratories to evaluate copy number changes or the mutational profile of this disease. Mitogenic stimulation of CLL specimens with CpG-oligonucleotide (CpG-ODN) has been identified as a reliable and reproducible means of obtaining a karyotype, facilitating a low-resolution genome-wide analysis. Across a cohort of 1255 CpG-ODN-stimulated CLL specimens, we describe the clinical utility associated with the combinatorial use of FISH and karyotyping. Our testing algorithm achieves a higher diagnostic yield (∼10%) through the detection of complex karyotypes, well-characterized chromosomal aberrations not covered by the traditional CLL FISH panel and through the detection of concurrent secondary malignancies. Moreover, the single cell nature of this approach permits the evaluation of emerging new clinical concepts including clonal dynamics and clonal evolution. This approach can be broadly applied by diagnostic laboratories to improve the utility of traditional and molecular cytogenetic studies of CLL. Am. J. Hematol. 91:978-983, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Adrian M. Dubuc
- Department of Pathology; Brigham and Women's Hospital; Boston Massachusetts
- Harvard Medical School; Boston Massachusetts
| | - Matthew S. Davids
- Harvard Medical School; Boston Massachusetts
- Department of Medical Oncology; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Mirela Pulluqi
- Department of Pathology; Brigham and Women's Hospital; Boston Massachusetts
| | - Olja Pulluqi
- Department of Pathology; Brigham and Women's Hospital; Boston Massachusetts
| | - Kevin Hoang
- Department of Medical Oncology; Dana-Farber Cancer Institute; Boston Massachusetts
| | | | - Cathy Schlich
- Department of Pathology; Brigham and Women's Hospital; Boston Massachusetts
| | - Jesus M. Hernández-Rivas
- IBSAL, IBMCC, Centro de Investigación del Cáncer, Universidad de Salamanca-CSIC; Salamanca 37007 Spain
- Department of Hematology; Hospital Universitario de Salamanca; Salamanca 37007 Spain
| | - Jennifer R. Brown
- Harvard Medical School; Boston Massachusetts
- Department of Medical Oncology; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Paola Dal Cin
- Department of Pathology; Brigham and Women's Hospital; Boston Massachusetts
- Harvard Medical School; Boston Massachusetts
| |
Collapse
|
187
|
Wiestner A. The role of B-cell receptor inhibitors in the treatment of patients with chronic lymphocytic leukemia. Haematologica 2016; 100:1495-507. [PMID: 26628631 DOI: 10.3324/haematol.2014.119123] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chronic lymphocytic leukemia is a malignancy of mature auto-reactive B cells. Genetic and functional studies implicate B-cell receptor signaling as a pivotal pathway in its pathogenesis. Full B-cell receptor activation requires tumor-microenvironment interactions in lymphoid tissues. Spleen tyrosine kinase, Bruton's tyrosine kinase, and the phosphatidylinositol 3-kinase (PI3K) δ isoform are essential for B-cell receptor signal transduction but also mediate the effect of other pathways engaged in chronic lymphocytic leukemia cells in the tissue-microenvironment. Orally bioavailable inhibitors of spleen tyrosine kinase, Bruton's tyrosine kinase, or PI3Kδ, induce high rates of durable responses. Ibrutinib, a covalent inhibitor of Bruton's tyrosine kinase, and idelalisib, a selective inhibitor of PI3Kδ, have obtained regulatory approval in chronic lymphocytic leukemia. Ibrutinib and idelalisib are active in patients with high-risk features, achieving superior disease control in difficult-to-treat patients than prior best therapy, making them the preferred agents for chronic lymphocytic leukemia with TP53 aberrations and for patients resistant to chemoimmunotherapy. In randomized trials, both ibrutinib, versus ofatumumab, and idelalisib in combination with rituximab, versus placebo with rituximab improved survival in relapsed/refractory chronic lymphocytic leukemia. Responses to B-cell receptor inhibitors are mostly partial, and within clinical trials treatment is continued until progression or occurrence of intolerable side effects. Ibrutinib and idelalisib are, overall, well tolerated; notable adverse events include increased bruising and incidence of atrial fibrillation on ibrutinib and colitis, pneumonitis and transaminase elevations on idelalisib. Randomized trials investigate the role of B-cell receptor inhibitors in first-line therapy and the benefit of combinations. This review discusses the biological basis for targeted therapy of chronic lymphocytic leukemia with B-cell receptor inhibitors, and summarizes the clinical experience with these agents.
Collapse
Affiliation(s)
- Adrian Wiestner
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
188
|
Stengel A, Kern W, Haferlach T, Meggendorfer M, Fasan A, Haferlach C. The impact of TP53 mutations and TP53 deletions on survival varies between AML, ALL, MDS and CLL: an analysis of 3307 cases. Leukemia 2016; 31:705-711. [PMID: 27680515 DOI: 10.1038/leu.2016.263] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/13/2016] [Accepted: 07/19/2016] [Indexed: 12/16/2022]
Abstract
Alterations in TP53 have been described in many cancer types including hematological neoplasms. We aimed at comparing TP53 mutations (mut) and deletions (del) in a large cohort of patients with hematological malignancies (n=3307), including AML (n=858), MDS (n=943), ALL (n=358), CLL (n=1148). Overall, alterations in TP53 were detected in 332/3307 cases (10%). The highest frequency was observed in ALL (total: 19%; mut+del: 6%; mut only: 8%; del only: 5%) and AML (total: 13%; mut+del: 5%; mut only: 7%; del only: 1%), whereas TP53 alterations occurred less frequently in CLL (total: 8%) and MDS (total: 7%). TP53 mutations were significantly more frequent in patients ⩾60 vs <60 years in AML (9% vs 2%, P<0.001) and ALL (12% vs 6%, P<0.001). TP53mut+del had a significant negative impact on overall survival in all entities, whereas differences were observed regarding TP53mut only or TP53del only: TP53mut only impacted survival in AML (36 vs 9 months, P<0.001) and MDS (65 vs 19 months, P<0.001), TP53del only in CLL (not reached vs 64 months, P=0.008) and MDS (65 vs 24 months, P=0.011). As substantial differences between the entities are observed regarding correlation to age and survival, we suggest evaluation of both TP53 deletion and mutation status.
Collapse
Affiliation(s)
- A Stengel
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - W Kern
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - T Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| | | | - A Fasan
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - C Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| |
Collapse
|
189
|
Le Bris Y, Struski S, Guièze R, Rouvellat C, Prade N, Troussard X, Tournilhac O, Béné MC, Delabesse E, Ysebaert L. Major prognostic value of complex karyotype in addition to TP53 and IGHV mutational status in first-line chronic lymphocytic leukemia. Hematol Oncol 2016; 35:664-670. [PMID: 27678008 DOI: 10.1002/hon.2349] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/03/2016] [Accepted: 08/05/2016] [Indexed: 11/05/2022]
Abstract
Chronic lymphocytic leukemia (CLL) is a lymphoproliferative disorder of remarkable heterogeneity as demonstrated by cytogenetics and molecular analyses. Complex karyotype (CK), TP53 deletions and/or mutations (TP53 disruption), IGVH mutational status, and, more recently, recurrent somatic mutations have been identified as prognostic markers in CLL. On a cohort of 110 patients with CLL treated with first-line fludarabin, cyclophosphamide, and rituximab treatment compared with 33 untreated (watch and wait) patients with CLL, we report more frequent complex karyotypes (34 vs 15%; P = .05), unmutated IGHV (70 vs 21%; P < .0001), ATM deletion (25 vs 6%, P = .02), and NOTCH mutation (3 vs 17%, P = .04). Among treated patients, 39 relapsed during the follow-up period. These patients were characterized before treatment by a higher incidence of trisomy 12 (38 vs 11%, P < .001) and TP53 disruption (31 vs 4%, P = .0002). A significantly shorter 5-year overall survival was found for treated patients with CK (72.4 vs 85.8%; P = .007), unmutated IGHV (70 vs 100%; P = .04), or TP53 disruption (55.7 vs 82.7%; P < .0001). Three risk groups were defined based on the status of TP53 disruption or unmutated IGVH, which differed significantly in terms of 5-year overall survival. Moreover, the presence of CK impacted pejoratively 5-year overall survival and progression-free survival in all these 3 groups. Conventional karyotyping therefore appears to be of value, CK being an additional factor, undetectable in classical FISH, in patients with CLL at the stage when therapy becomes required.
Collapse
Affiliation(s)
- Yannick Le Bris
- Laboratoire d'Hématologie, CHU de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France.,Service d'Hématologie Biologique, CHU de Nantes, Nantes, France
| | - Stéphanie Struski
- Laboratoire d'Hématologie, CHU de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Romain Guièze
- Service d'Hématologie Clinique, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Caroline Rouvellat
- Laboratoire d'Hématologie, CHU de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Naïs Prade
- Laboratoire d'Hématologie, CHU de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | | | - Olivier Tournilhac
- Service d'Hématologie Clinique, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Marie C Béné
- Service d'Hématologie Biologique, CHU de Nantes, Nantes, France
| | - Eric Delabesse
- Laboratoire d'Hématologie, CHU de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Loïc Ysebaert
- Service d'Hématologie Clinique, CHU de Toulouse, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| |
Collapse
|
190
|
Extensive next-generation sequencing analysis in chronic lymphocytic leukemia at diagnosis: clinical and biological correlations. J Hematol Oncol 2016; 9:88. [PMID: 27633522 PMCID: PMC5025606 DOI: 10.1186/s13045-016-0320-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 09/06/2016] [Indexed: 11/10/2022] Open
Abstract
Background In chronic lymphocytic leukemia (CLL), next-generation sequencing (NGS) analysis represents a sensitive, reproducible, and resource-efficient technique for routine screening of gene mutations. Methods We performed an extensive biologic characterization of newly diagnosed CLL, including NGS analysis of 20 genes frequently mutated in CLL and karyotype analysis to assess whether NGS and karyotype results could be of clinical relevance in the refinement of prognosis and assessment of risk of progression. The genomic DNA from peripheral blood samples of 200 consecutive CLL patients was analyzed using Ion Torrent Personal Genome Machine, a NGS platform that uses semiconductor sequencing technology. Karyotype analysis was performed using efficient mitogens. Results Mutations were detected in 42.0 % of cases with 42.8 % of mutated patients presenting 2 or more mutations. The presence of mutations by NGS was associated with unmutated IGHV gene (p = 0.009), CD38 positivity (p = 0.010), risk stratification by fluorescence in situ hybridization (FISH) (p < 0.001), and the complex karyotype (p = 0.003). A high risk as assessed by FISH analysis was associated with mutations affecting TP53 (p = 0.012), BIRC3 (p = 0.003), and FBXW7 (p = 0.003) while the complex karyotype was significantly associated with TP53, ATM, and MYD88 mutations (p = 0.003, 0.018, and 0.001, respectively). By multivariate analysis, the multi-hit profile (≥2 mutations by NGS) was independently associated with a shorter time to first treatment (p = 0.004) along with TP53 disruption (p = 0.040), IGHV unmutated status (p < 0.001), and advanced stage (p < 0.001). Advanced stage (p = 0.010), TP53 disruption (p < 0.001), IGHV unmutated status (p = 0.020), and the complex karyotype (p = 0.007) were independently associated with a shorter overall survival. Conclusions At diagnosis, an extensive biologic characterization including NGS and karyotype analyses using novel mitogens may offer new perspectives for a better refinement of risk stratification that could be of help in the clinical management of CLL patients. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0320-z) contains supplementary material, which is available to authorized users.
Collapse
|
191
|
Abstract
High-throughput sequencing of cancer genomes is increasingly becoming an essential tool of clinical oncology that facilitates target identification and targeted therapy within the context of precision medicine. The cumulative profiles of somatic mutations in cancer yielded by comprehensive molecular studies also constitute a fingerprint of historical exposures to exogenous and endogenous mutagens, providing insight into cancer evolution and etiology. Mutational signatures that were first established by inspection of the TP53 gene somatic landscape have now been confirmed and expanded by comprehensive sequencing studies. Further, the degree of granularity achieved by deep sequencing allows detection of low-abundance mutations with clinical relevance. In tumors, they represent the emergence of small aggressive clones; in normal tissues, they signal a mutagenic exposure related to cancer risk; and, in blood, they may soon become effective surveillance tools for diagnostic purposes and for monitoring of cancer prognosis and recurrence.
Collapse
Affiliation(s)
- Ana I Robles
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Jin Jen
- Department of Laboratory Medicine and Pathology, Division of Experimental Pathology, and Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
192
|
Amin NA, Balasubramanian S, Saiya-Cork K, Shedden K, Hu N, Malek SN. Cell-Intrinsic Determinants of Ibrutinib-Induced Apoptosis in Chronic Lymphocytic Leukemia. Clin Cancer Res 2016; 23:1049-1059. [PMID: 27535981 DOI: 10.1158/1078-0432.ccr-15-2921] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 07/07/2016] [Accepted: 08/08/2016] [Indexed: 01/18/2023]
Abstract
Purpose: Ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor, is approved for the treatment of relapsed chronic lymphocytic leukemia (CLL) and CLL with del17p. Mechanistically, ibrutinib interferes with B-cell receptor (BCR) signaling as well as multiple CLL cell-to-microenvironment interactions. Given the importance of ibrutinib in the management of CLL, a deeper understanding of factors governing sensitivity and resistance is warranted.Experimental Design: We studied 48 longitudinally sampled paired CLL samples, 42 of which were procured before and after standard CLL chemotherapies, and characterized them for well-studied CLL molecular traits as well as by whole-exome sequencing and SNP 6.0 array profiling. We exposed these samples to 0.25 to 5 μmol/L of ibrutinib ex vivo and measured apoptosis fractions as well as BCR signaling by immunoblotting. We disrupted TP53 in HG3, PGA1, and PG-EBV cell lines and measured BCR signaling and ibrutinib responses.Results: CLL samples demonstrated a surprisingly wide range of ex vivo sensitivities to ibrutinib, with IC50 values ranging from 0.4 to 9.7 μmol/L. Unmutated IGVH status, elevated ZAP70 expression, and trisomy 12 were associated with heightened sensitivity to ibrutinib treatment. Five CLL samples were substantially more resistant to ibrutinib following relapse from chemotherapy; of these, three had acquired a del17p/TP53-mutated status. A validation sample of 15 CLL carrying TP53 mutations, of which 13 carried both del17p and a TP53 mutation, confirmed substantially less sensitivity to ibrutinib-induced apoptosis.Conclusions: This study identifies that CLL harboring del17p/TP53-mutated cells are substantially less sensitive to ibrutinib-induced apoptosis than del17p/TP53 wild-type cells. Clin Cancer Res; 23(4); 1049-59. ©2016 AACR.
Collapse
Affiliation(s)
- Nisar A Amin
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | | | - Kamlai Saiya-Cork
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Kerby Shedden
- Department of Statistics, University of Michigan, Ann Arbor, Michigan
| | - Nan Hu
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Sami N Malek
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
193
|
TP53 dysfunction in CLL: Implications for prognosis and treatment. Best Pract Res Clin Haematol 2016; 29:90-99. [PMID: 27742075 DOI: 10.1016/j.beha.2016.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 07/26/2016] [Accepted: 08/04/2016] [Indexed: 02/01/2023]
Abstract
Despite the availability of novel targeted agents, TP53 defects remain the most important adverse prognostic factor in chronic lymphocytic leukemia (CLL). Detection of deletion of TP53 locus (17p deletion) by fluorescent in situ hybridization (FISH) has become standard and performed prior to every line of treatment as the incidence dramatically increases as relapses occur. As monoallelic mutations of TP53 equally affect outcome, novel methods are being developed to improve detection of TP53 defects and include next-generation sequencing (NGS) and functional assays. TP53 defects highly affect outcome of immunochemotherapy but also alter response durations of tyrosine kinase inhibitors. Although BCR-targeting agents and Bcl-2-inhibitos have achieved durable responses in some patients with TP53 defects, long-term follow-up is currently lacking. In this review biological and clinical consequences of TP53 dysfunction as well as applicability of currently available methods to detect TP53 defects are described. In addition, proposed novel therapeutic strategies specifically for patients with TP53 dysfunction are discussed. In summary, the only curative treatment option for TP53-defective CLL is still allogeneic hematopoietic stem cell transplantation. Other treatment strategies such as rationale combinations of agents with different (TP53 independent) targets, including kinase inhibitors and inhibitors of anti-apoptotic molecules but also immunomodulatory agents need to be further explored.
Collapse
|
194
|
|
195
|
Yu L, Kim HT, Kasar S, Benien P, Du W, Hoang K, Aw A, Tesar B, Improgo R, Fernandes S, Radhakrishnan S, Klitgaard J, Lee C, Getz G, Setlur SR, Brown JR. Survival of Del17p CLL Depends on Genomic Complexity and Somatic Mutation. Clin Cancer Res 2016; 23:735-745. [PMID: 27503198 DOI: 10.1158/1078-0432.ccr-16-0594] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/17/2016] [Accepted: 07/17/2016] [Indexed: 12/30/2022]
Abstract
PURPOSE Chronic lymphocytic leukemia (CLL) with 17p deletion typically progresses quickly and is refractory to most conventional therapies. However, some del(17p) patients do not progress for years, suggesting that del(17p) is not the only driving event in CLL progression. We hypothesize that other concomitant genetic abnormalities underlie the clinical heterogeneity of del(17p) CLL. EXPERIMENTAL DESIGN We profiled the somatic mutations and copy number alterations (CNA) in a large group of del(17p) CLLs as well as wild-type CLL and analyzed the genetic basis of their clinical heterogeneity. RESULTS We found that increased somatic mutation number associates with poor overall survival independent of 17p deletion (P = 0.003). TP53 mutation was present in 81% of del(17p) CLL, mostly clonal (82%), and clonal mutations with del(17p) exhibit shorter overall survival than subclonal mutations with del(17p) (P = 0.019). Del(17p) CLL has a unique driver mutation profile, including NOTCH1 (15%), RPS15 (12%), DDX3X (8%), and GPS2 (6%). We found that about half of del(17p) CLL cases have recurrent deletions at 3p, 4p, or 9p and that any of these deletions significantly predicts shorter overall survival. In addition, the number of CNAs, but not somatic mutations, predicts shorter time to treatment among patients untreated at sampling. Indolent del(17p) CLLs were characterized by absent or subclonal TP53 mutation and few CNAs, with no difference in somatic mutation number. CONCLUSIONS We conclude that del(17p) has a unique genomic profile and that clonal TP53 mutations, 3p, 4p, or 9p deletions, and genomic complexity are associated with shorter overall survival. Clin Cancer Res; 23(3); 735-45. ©2016 AACR.
Collapse
Affiliation(s)
- Lijian Yu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Haesook T Kim
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA
| | - Siddha Kasar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | | | - Wei Du
- Brigham and Women's Hospital, Boston, MA
| | - Kevin Hoang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Andrew Aw
- Division of Hematology, The Ottawa Hospital, Ottawa, Ontario
| | - Bethany Tesar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Reina Improgo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Stacey Fernandes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Josephine Klitgaard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Charles Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT
| | - Gad Getz
- Broad Institute of Harvard and MIT, Cambridge, MA.,Massachusetts General Hospital Cancer Center and Department of Pathology, Boston, MA
| | - Sunita R Setlur
- Brigham and Women's Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Jennifer R Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
196
|
Intratumoral genetic heterogeneity and number of cytogenetic aberrations provide additional prognostic significance in chronic lymphocytic leukemia. Genet Med 2016; 19:182-191. [PMID: 27467457 DOI: 10.1038/gim.2016.81] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 05/03/2016] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Chronic lymphocytic leukemia (CLL) is a heterogeneous disease with cytogenetic aberrations that are still considered the gold standard of prognostic factors. However, heterogeneity remains within each cytogenetic group, especially in patients with concomitant cytogenetic aberrations. METHODS A panel of DNA probes was used to detect cytogenetic aberrations, including RB1/D13S25 at 13q14, ATM at 11q22, TP53 at 17p13, CEP12 and IGH translocation at 14q32, by fluorescence in situ hybridization. A comprehensive method integrating the number of cytogenetic aberrations and intratumoral genetic heterogeneity was used to analyze the prognosis for patients with concomitant aberrations. RESULTS Within the conventional favorable or neutral prognostic groups (i.e., with del 13q, trisomy 12, and/or t(14q32)), the coincidence of these three aberrations worsened survival in terms of time to first therapy, progression-free survival, and overall survival. However, within the conventional unfavorable prognostic group (i.e., del 11q or del 17p), patients with a minor unfavorable clone had an unexpected survival advantage compared with patients with a major unfavorable clone. A new cytogenetic prognostic system that integrates the number of cytogenetic aberrations and intratumoral genetic subclones was more precise than the conventional system. CONCLUSION The number of cytogenetic aberrations and the size of intratumoral genetic subclones should be comprehensively considered to determine the prognosis for CLL.Genet Med 19 2, 182-191.
Collapse
|
197
|
Gordon MJ, Raess PW, Young K, Spurgeon SEF, Danilov AV. Ibrutinib is an effective treatment for B-cell prolymphocytic leukaemia. Br J Haematol 2016; 179:501-503. [DOI: 10.1111/bjh.14224] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Max J. Gordon
- Department of Medicine; Oregon Health and Science University; Portland OR USA
| | - Philipp W. Raess
- Department of Pathology; Oregon Health and Science University; Portland OR USA
| | - Kathleen Young
- Department of Medicine; Oregon Health and Science University; Portland OR USA
| | | | - Alexey V. Danilov
- Department of Medicine; Oregon Health and Science University; Portland OR USA
| |
Collapse
|
198
|
|
199
|
Lazarian G, Tausch E, Eclache V, Sebaa A, Bianchi V, Letestu R, Collon JF, Lefebvre V, Gardano L, Varin-Blank N, Soussi T, Stilgenbauer S, Cymbalista F, Baran-Marszak F. TP53 mutations are early events in chronic lymphocytic leukemia disease progression and precede evolution to complex karyotypes. Int J Cancer 2016; 139:1759-63. [PMID: 27270786 DOI: 10.1002/ijc.30222] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 05/18/2016] [Accepted: 05/25/2016] [Indexed: 01/08/2023]
Abstract
TP53 abnormalities lead to resistance to purine analogues and are found in over 40% of patients with refractory chronic lymphocytic leukemia (CLL). At diagnosis, no more than 5% of patients carry the 17p deletion, most cases harbour mutations within the other TP53 allele. The incidence of a TP53 mutation as the only alteration is approximately 5%, but this depends on the sensitivity of the technique. Recently, having a complex karyotype has been considered a strong adverse prognostic factor. However, there are no longitudinal studies simultaneously examining the presence of the 17p deletion, TP53 mutations and karyotype abnormalities. We conducted a retrospective longitudinal study of 31 relapsed/refractory CLL patients. Two to six blood samples per patient were analyzed, with a median follow-up of 8 years. In this report, we assessed the sequence of events of TP53 clonal evolution and correlated the presence of TP53 abnormalities to genetic instability during progression and treatment. Next-generation sequencing allowed the early detection of TP53 mutated clones and was able to be performed on a routine basis, demonstrating an excellent correlation between the Illumina and Ion Torrent technologies. We concluded that TP53 mutations are early events and precede clonal evolution to complex karyotypes. We strongly recommend the early and iterated detection of TP53 mutations in progressive cases.
Collapse
Affiliation(s)
- Gregory Lazarian
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Labex Inflamex, Université Paris 13, Sorbonne Paris Cité, Bobigny, France.,Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Eugen Tausch
- Department of Internal Medicine III, Ulm University, Ulm, Germany
| | - Virginie Eclache
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Amel Sebaa
- Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Vincent Bianchi
- Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Remi Letestu
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Labex Inflamex, Université Paris 13, Sorbonne Paris Cité, Bobigny, France.,Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Jean-Francois Collon
- Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Valerie Lefebvre
- Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Laura Gardano
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Labex Inflamex, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - Nadine Varin-Blank
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Labex Inflamex, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - Thierry Soussi
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska (CCK) R8:04, Stockholm SE-171 76, Sweden; Sorbonne Universités, UPMC Univ Paris 06, Paris, F-75005, France.,INSERM, U1138, Centre de Recherche des Cordeliers, Paris, France and Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Florence Cymbalista
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Labex Inflamex, Université Paris 13, Sorbonne Paris Cité, Bobigny, France.,Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| | - Fanny Baran-Marszak
- U978 Institut National De La Santé Et De La Recherche Médicale, Bobigny, France.,Labex Inflamex, Université Paris 13, Sorbonne Paris Cité, Bobigny, France.,Hôpital Avicenne, Assistance Publique-Hôpitaux De Paris, Bobigny, Service D'Hématologie Biologique, France
| |
Collapse
|
200
|
Kojima K, Ishizawa J, Andreeff M. Pharmacological activation of wild-type p53 in the therapy of leukemia. Exp Hematol 2016; 44:791-798. [PMID: 27327543 DOI: 10.1016/j.exphem.2016.05.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
The tumor suppressor p53 is inactivated by mutations in the majority of human solid tumors. Conversely, p53 mutations are rare in leukemias and are only observed in a small fraction of the patient population, predominately in patients with complex karyotype acute myeloid leukemia or hypodiploid acute lymphoblastic leukemia. However, the loss of p53 function in leukemic cells is often caused by abnormalities in p53-regulatory proteins, including overexpression of MDM2/MDMX, deletion of CDKN2A/ARF, and alterations in ATM. For example, MDM2 inhibits p53-mediated transcription, promotes its nuclear export, and induces proteasome-dependent degradation. The MDM2 homolog MDMX is another direct regulator of p53 that inhibits p53-mediated transcription. Several small-molecule inhibitors and stapled peptides targeting MDM2 and MDMX have been developed and have recently entered clinical trials. The clinical trial results of the first clinically used MDM2 inhibitor, RG7112, illustrated promising p53 activation and apoptosis induction in leukemia cells as proof of concept. Side effects of RG7112 were most prominent in suppression of thrombopoiesis and gastrointestinal symptoms in leukemia patients. Predictive biomarkers for response to MDM2 inhibitors have been proposed, but they require further validation both in vitro and in vivo so that the accumulated knowledge concerning pathological p53 dysregulation in leukemia and novel molecular-targeted strategies to overcome this dysregulation can be translated safely and efficiently into novel clinical therapeutics.
Collapse
Affiliation(s)
- Kensuke Kojima
- Department of Hematology, Respiratory Medicine and Oncology, Division of Medicine, Saga University, Saga, Japan; Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.
| | - Jo Ishizawa
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Section of Molecular Hematology and Therapy, Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|