251
|
Kantorova B, Malcikova J, Smardova J, Pavlova S, Trbusek M, Tom N, Plevova K, Tichy B, Truong S, Diviskova E, Kotaskova J, Oltova A, Patten N, Brychtova Y, Doubek M, Mayer J, Pospisilova S. TP53 mutation analysis in chronic lymphocytic leukemia: comparison of different detection methods. Tumour Biol 2014; 36:3371-80. [PMID: 25527155 DOI: 10.1007/s13277-014-2971-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/10/2014] [Indexed: 11/24/2022] Open
Abstract
TP53 gene defects represent a strong adverse prognostic factor for patient survival and treatment resistance in chronic lymphocytic leukemia (CLL). Although various methods for TP53 mutation analysis have been reported, none of them allow the identification of all occurring sequence variants, and the most suitable methodology is still being discussed. The aim of this study was to determine the limitations of commonly used methods for TP53 mutation examination in CLL and propose an optimal approach for their detection. We examined 182 CLL patients enriched for high-risk cases using denaturing high-performance liquid chromatography (DHPLC), functional analysis of separated alleles in yeast (FASAY), and the AmpliChip p53 Research Test in parallel. The presence of T53 gene mutations was also evaluated using ultra-deep next generation sequencing (NGS) in 69 patients. In total, 79 TP53 mutations in 57 (31 %) patients were found; among them, missense substitutions predominated (68 % of detected mutations). Comparing the efficacy of the methods used, DHPLC and FASAY both combined with direct Sanger sequencing achieved the best results, identifying 95 % and 93 % of TP53-mutated patients. Nevertheless, we showed that in CLL patients carrying low-proportion TP53 mutation, the more sensitive approach, e.g., ultra-deep NGS, might be more appropriate. TP53 gene analysis using DHPLC or FASAY is a suitable approach for mutation detection. Ultra-deep NGS has the potential to overcome shortcomings of methods currently used, allows the detection of minor proportion mutations, and represents thus a promising methodology for near future.
Collapse
Affiliation(s)
- Barbara Kantorova
- Central European Institute of Technology (CEITEC), Masaryk University Brno, Kamenice 5, CZ-625 00, Brno, Czech Republic
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
252
|
Dreger P, Schetelig J, Andersen N, Corradini P, van Gelder M, Gribben J, Kimby E, Michallet M, Moreno C, Stilgenbauer S, Montserrat E. Managing high-risk CLL during transition to a new treatment era: stem cell transplantation or novel agents? Blood 2014; 124:3841-9. [PMID: 25301705 PMCID: PMC4276025 DOI: 10.1182/blood-2014-07-586826] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/27/2014] [Indexed: 01/01/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) has been considered as the treatment of choice for patients with high-risk chronic lymphocytic leukemia (HR-CLL; ie, refractory to purine analogs, short response [<24 months] to chemoimmunotherapy, and/or presence of del[17p]/TP53 mutations). Currently, treatment algorithms for HR-CLL are being challenged by the introduction of novel classes of drugs. Among them, BCR signal inhibitors (BCRi) and B-cell lymphoma 2 antagonists (BCL2a) appear particularly promising. As a result of the growing body of favorable outcome data reported for BCRi/BCL2a, uncertainty is emerging on how to advise patients with HR-CLL about indication for and timing of HSCT. This article provides an overview of currently available evidence and theoretical considerations to guide this difficult decision process. Until the risks and benefits of different treatment strategies are settled, all patients with HR-CLL should be considered for treatment with BCRi/BCL2a. For patients who respond to these agents, there are 2 treatment possibilities: (1) performing an HSCT or (2) continuing treatment with the novel drug. Individual disease-specific and transplant-related risk factors, along with patient's preferences, should be taken into account when recommending one of these treatments over the other.
Collapse
Affiliation(s)
- Peter Dreger
- Department Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Johannes Schetelig
- Department Medicine I, University of Dresden, Dresden, Germany; German Bone Marrow Donor Registry, Tübingen, Germany
| | - Niels Andersen
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Paolo Corradini
- Department Hematology and Pediatric Onco-Hematology, Istituto Nazionale dei Tumori, Milan, Italy
| | - Michel van Gelder
- Department Internal Medicine/Hematology, Maastricht University, Maastricht, The Netherlands
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Eva Kimby
- Department of Medicine/Hematology Unit, Karolinska Institute, Huddinge, Sweden
| | | | - Carol Moreno
- Hospital de la Santa Creu Sant Pau, Barcelona, Spain
| | | | - Emili Montserrat
- Department of Hematology, Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
253
|
Wang J, Khiabanian H, Rossi D, Fabbri G, Gattei V, Forconi F, Laurenti L, Marasca R, Del Poeta G, Foà R, Pasqualucci L, Gaidano G, Rabadan R. Tumor evolutionary directed graphs and the history of chronic lymphocytic leukemia. eLife 2014; 3. [PMID: 25496728 PMCID: PMC4308685 DOI: 10.7554/elife.02869] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022] Open
Abstract
Cancer is a clonal evolutionary process, caused by successive accumulation of genetic alterations providing milestones of tumor initiation, progression, dissemination, and/or resistance to certain therapeutic regimes. To unravel these milestones we propose a framework, tumor evolutionary directed graphs (TEDG), which is able to characterize the history of genetic alterations by integrating longitudinal and cross-sectional genomic data. We applied TEDG to a chronic lymphocytic leukemia (CLL) cohort of 70 patients spanning 12 years and show that: (a) the evolution of CLL follows a time-ordered process represented as a global flow in TEDG that proceeds from initiating events to late events; (b) there are two distinct and mutually exclusive evolutionary paths of CLL evolution; (c) higher fitness clones are present in later stages of the disease, indicating a progressive clonal replacement with more aggressive clones. Our results suggest that TEDG may constitute an effective framework to recapitulate the evolutionary history of tumors. DOI:http://dx.doi.org/10.7554/eLife.02869.001 A historical event is often the culmination of the preceding circumstances. The same can be said of cancer as a disease. Cancer results from genetic mutations that disrupt the normal biological processes within a cell, removing the fail-safes that prevent it from growing and reproducing uncontrollably. Cancer is not caused by just one mutation, and once one gene is malfunctioning, other genes become much more likely to mutate. Although modern sequencing methods have revealed many of the genes that mutate in several different kinds of cancer, uncovering when each of these mutations occurs has been more difficult. Knowing when each mutation occurs could make it easier to predict how the cancer will progress and could also help target cancer treatments more effectively. Wang, Khiabanian, Rossi et al. have devised a new method of studying the history of genetic mutations of cancer patients. This combines a ‘longitudinal’ method that looks at how mutations develop in a single tumor by taking samples from it at different times and ‘cross-sectional’ methods that make predictions based on data collected from a large number of patients. Wang, Khiabanian, Rossi et al. call this method ‘tumor evolutionary directed graphs’ (TEDG), as it produces a graph that shows how different gene mutations are related to each other. Initial tests showed that the TEDG method could accurately decipher the main chain of events in cancer evolution when used on data collected from at least 30 patients. Wang, Khiabanian, Rossi et al. then used TEDG on data from 164 tumor samples collected over 12 years from 70 patients with chronic lymphocytic leukemia, the type of leukemia that is most widespread amongst adults in Western countries. This uncovered two separate ways that this cancer may develop, one of which has a higher risk of life-threatening complications. Knowing which of the two ways chronic lymphocytic leukemia is progressing in a patient could help treat the disease, as each pathway responds differently to different treatments. In addition, understanding the paths that cancer progression follows could also provide early warning signals of the mutations that will occur next. This could help to develop alternative, targeted cancer treatments. DOI:http://dx.doi.org/10.7554/eLife.02869.002
Collapse
Affiliation(s)
- Jiguang Wang
- Department of Biomedical Informatics, Columbia University, New York, United States
| | - Hossein Khiabanian
- Department of Biomedical Informatics, Columbia University, New York, United States
| | - Davide Rossi
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Giulia Fabbri
- Institute for Cancer Genetics, Columbia University, New York, United States
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico, Aviano, Italy
| | - Francesco Forconi
- Cancer Sciences Unit, Cancer Research UK Centre, University of Southampton, Southampton, United Kingdom
| | - Luca Laurenti
- Institute of Hematology, Catholic University of the Sacred Heart, Rome, Italy
| | - Roberto Marasca
- Division of Hematology, Department of Oncology and Hematology, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Robin Foà
- Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, United States
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, Amedeo Avogadro University of Eastern Piedmont, Novara, Italy
| | - Raul Rabadan
- Department of Biomedical Informatics, Columbia University, New York, United States
| |
Collapse
|
254
|
Strefford JC. The genomic landscape of chronic lymphocytic leukaemia: biological and clinical implications. Br J Haematol 2014; 169:14-31. [PMID: 25496136 DOI: 10.1111/bjh.13254] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic lymphocytic leukaemia (CLL) remains at the forefront of the genetic analysis of human tumours, principally due its prevalence, protracted natural history and accessibility to suitable material for analysis. With the application of high-throughput genetic technologies, we have an unbridled view of the architecture of the CLL genome, including a comprehensive description of the copy number and mutational landscape of the disease, a detailed picture of clonal evolution during pathogenesis, and the molecular mechanisms that drive genomic instability and therapeutic resistance. This work has nuanced the prognostic importance of established copy number alterations, and identified novel prognostically relevant gene mutations that function within biological pathways that are attractive treatment targets. Herein, an overview of recent genomic discoveries will be reviewed, with associated biological and clinical implications, and a view into how clinical implementation may be facilitated.
Collapse
Affiliation(s)
- Jonathan C Strefford
- Cancer Genomics, Academic Unit of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
255
|
Sutton LA, Ljungström V, Mansouri L, Young E, Cortese D, Navrkalova V, Malcikova J, Muggen AF, Trbusek M, Panagiotidis P, Davi F, Belessi C, Langerak AW, Ghia P, Pospisilova S, Stamatopoulos K, Rosenquist R. Targeted next-generation sequencing in chronic lymphocytic leukemia: a high-throughput yet tailored approach will facilitate implementation in a clinical setting. Haematologica 2014; 100:370-6. [PMID: 25480502 DOI: 10.3324/haematol.2014.109777] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Next-generation sequencing has revealed novel recurrent mutations in chronic lymphocytic leukemia, particularly in patients with aggressive disease. Here, we explored targeted re-sequencing as a novel strategy to assess the mutation status of genes with prognostic potential. To this end, we utilized HaloPlex targeted enrichment technology and designed a panel including nine genes: ATM, BIRC3, MYD88, NOTCH1, SF3B1 and TP53, which have been linked to the prognosis of chronic lymphocytic leukemia, and KLHL6, POT1 and XPO1, which are less characterized but were found to be recurrently mutated in various sequencing studies. A total of 188 chronic lymphocytic leukemia patients with poor prognostic features (unmutated IGHV, n=137; IGHV3-21 subset #2, n=51) were sequenced on the HiSeq 2000 and data were analyzed using well-established bioinformatics tools. Using a conservative cutoff of 10% for the mutant allele, we found that 114/180 (63%) patients carried at least one mutation, with mutations in ATM, BIRC3, NOTCH1, SF3B1 and TP53 accounting for 149/177 (84%) of all mutations. We selected 155 mutations for Sanger validation (variant allele frequency, 10-99%) and 93% (144/155) of mutations were confirmed; notably, all 11 discordant variants had a variant allele frequency between 11-27%, hence at the detection limit of conventional Sanger sequencing. Technical precision was assessed by repeating the entire HaloPlex procedure for 63 patients; concordance was found for 77/82 (94%) mutations. In summary, this study demonstrates that targeted next-generation sequencing is an accurate and reproducible technique potentially suitable for routine screening, eventually as a stand-alone test without the need for confirmation by Sanger sequencing.
Collapse
Affiliation(s)
- Lesley-Ann Sutton
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Viktor Ljungström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Larry Mansouri
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Emma Young
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Diego Cortese
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| | - Veronika Navrkalova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jitka Malcikova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Alice F Muggen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Martin Trbusek
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | | | - Frederic Davi
- Laboratory of Hematology and Universite Pierre et Marie Curie, Hopital Pitie-Salpetriere, Paris, France
| | | | - Anton W Langerak
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, The Netherlands
| | - Paolo Ghia
- Università Vita-Salute San Raffaele, Milan, Italy Division of Molecular Oncology and Department of Onco-Hematology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sarka Pospisilova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Kostas Stamatopoulos
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden Institute of Applied Biosciences, CERTH, Thessaloniki, Greece Hematology Department and HCT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Richard Rosenquist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Sweden
| |
Collapse
|
256
|
Zent CS, Burack WR. Mutations in chronic lymphocytic leukemia and how they affect therapy choice: focus on NOTCH1, SF3B1, and TP53. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2014; 2014:119-124. [PMID: 25696844 DOI: 10.1182/asheducation-2014.1.119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by a relatively small number of recurrent genetic defects. These can be evaluated by clinically available methods such as fluorescent in situ hybridization and targeted sequencing approaches to provide data that can be very helpful in prognostication and planning of treatment. Acquired defects in the p53 pathway, activating mutations of NOTCH1, and dysfunctional mutations of SF3B1 and BIRC3 identify patients with higher risk of progressive disease, poorer responses to conventional chemoimmunotherapy, and shorter survival. Risk stratification using these data can identify patients with aggressive CLL who require careful monitoring and are unlikely to have durable responses to chemoimmunotherapy at disease progression. Patients with defective DNA damage repair mechanisms because of p53 dysfunction should be considered for non-chemotherapy-based regimens including tyrosine kinase inhibitors, BCL2 inhibitors, monoclonal antibodies, and immunological therapies including allogeneic transplantation and chimeric antigen receptor-targeted T cells. Conversely, patients with no high-risk mutations can usually be monitored for a prolonged time and are likely to have durable responses to chemoimmunotherapy at disease progression. New technologies for genetic analysis such as targeted next-generation sequencing have the potential to make these analyses cheaper, faster, and more widely available. Comprehensive genetic analysis of patients both at diagnosis and before treatment for progressive disease could become an integral component of care for CLL.
Collapse
MESH Headings
- Clonal Evolution
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology
- Mutation/genetics
- Receptor, Notch1/genetics
- Ribonucleoprotein, U2 Small Nuclear/genetics
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Clive S Zent
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - W Richard Burack
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
257
|
Paíno T, Paiva B, Sayagués JM, Mota I, Carvalheiro T, Corchete LA, Aires-Mejía I, Pérez JJ, Sanchez ML, Barcena P, Ocio EM, San-Segundo L, Sarasquete ME, García-Sanz R, Vidriales MB, Oriol A, Hernández MT, Echeveste MA, Paiva A, Blade J, Lahuerta JJ, Orfao A, Mateos MV, Gutiérrez NC, San-Miguel JF. Phenotypic identification of subclones in multiple myeloma with different chemoresistant, cytogenetic and clonogenic potential. Leukemia 2014; 29:1186-94. [DOI: 10.1038/leu.2014.321] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 10/31/2014] [Accepted: 11/04/2014] [Indexed: 01/06/2023]
|
258
|
Bacher U, Kohlmann A, Haferlach T. Mutational profiling in patients with MDS: ready for every-day use in the clinic? Best Pract Res Clin Haematol 2014; 28:32-42. [PMID: 25659728 DOI: 10.1016/j.beha.2014.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 11/04/2014] [Indexed: 12/18/2022]
Abstract
Multiple recurrent somatic mutations were identified in the majority of patients with myelodysplastic syndromes (MDS), but investigating the broad spectrum of molecular markers in MDS exceeds many laboratories' capacity when traditional molecular techniques are used. High-throughput second generation sequencing (=next-generation sequencing, NGS) has proven to be applicable for comprehensive biomarker mutation analyses allowing to increase diagnostic sensitivity and accuracy and to improve risk stratification and prognostication in addition to cytomorphology and cytogenetic analysis in patients with MDS. Amplicon deep-sequencing enables comprehensive biomarker analysis in a multitude of patients per investigation in an acceptable turn-around time and at affordable costs. Comprehensive myeloid marker panels were successfully introduced into diagnostic practice. Therefore, molecular mutation analysis is ready for use in all patients with suspected MDS, may contribute to risk stratification in possible candidates for allogeneic stem cell transplantation, and should become an integral part of clinical research studies in MDS patients.
Collapse
|
259
|
Deep sequencing identifies genetic heterogeneity and recurrent convergent evolution in chronic lymphocytic leukemia. Blood 2014; 125:492-8. [PMID: 25377784 DOI: 10.1182/blood-2014-06-580563] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Recent high-throughput sequencing and microarray studies have characterized the genetic landscape and clonal complexity of chronic lymphocytic leukemia (CLL). Here, we performed a longitudinal study in a homogeneously treated cohort of 12 patients, with sequential samples obtained at comparable stages of disease. We identified clonal competition between 2 or more genetic subclones in 70% of the patients with relapse, and stable clonal dynamics in the remaining 30%. By deep sequencing, we identified a high reservoir of genetic heterogeneity in the form of several driver genes mutated in small subclones underlying the disease course. Furthermore, in 2 patients, we identified convergent evolution, characterized by the combination of genetic lesions affecting the same genes or copy number abnormality in different subclones. The phenomenon affects multiple CLL putative driver abnormalities, including mutations in NOTCH1, SF3B1, DDX3X, and del(11q23). This is the first report documenting convergent evolution as a recurrent event in the CLL genome. Furthermore, this finding suggests the selective advantage of specific combinations of genetic lesions for CLL pathogenesis in a subset of patients.
Collapse
|
260
|
Malcikova J, Stano-Kozubik K, Tichy B, Kantorova B, Pavlova S, Tom N, Radova L, Smardova J, Pardy F, Doubek M, Brychtova Y, Mraz M, Plevova K, Diviskova E, Oltova A, Mayer J, Pospisilova S, Trbusek M. Detailed analysis of therapy-driven clonal evolution of TP53 mutations in chronic lymphocytic leukemia. Leukemia 2014; 29:877-85. [PMID: 25287991 PMCID: PMC4396398 DOI: 10.1038/leu.2014.297] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 09/17/2014] [Accepted: 09/18/2014] [Indexed: 11/28/2022]
Abstract
In chronic lymphocytic leukemia (CLL), the worst prognosis is associated with TP53 defects with the affected patients being potentially directed to alternative treatment. Therapy administration was shown to drive the selection of new TP53 mutations in CLL. Using ultra-deep next-generation sequencing (NGS), we performed a detailed analysis of TP53 mutations' clonal evolution. We retrospectively analyzed samples that were assessed as TP53-wild-type (wt) by FASAY from 20 patients with a new TP53 mutation detected in relapse and 40 patients remaining TP53-wt in relapse. Minor TP53-mutated subclones were disclosed in 18/20 patients experiencing later mutation selection, while only one minor-clone mutation was observed in those patients remaining TP53-wt (n=40). We documented that (i) minor TP53 mutations may be present before therapy and may occur in any relapse; (ii) the majority of TP53-mutated minor clones expand to dominant clone under the selective pressure of chemotherapy, while persistence of minor-clone mutations is rare; (iii) multiple minor-clone TP53 mutations are common and may simultaneously expand. In conclusion, patients with minor-clone TP53 mutations carry a high risk of mutation selection by therapy. Deep sequencing can shift TP53 mutation identification to a period before therapy administration, which might be of particular importance for clinical trials.
Collapse
Affiliation(s)
- J Malcikova
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - K Stano-Kozubik
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - B Tichy
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - B Kantorova
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - S Pavlova
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - N Tom
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - L Radova
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - J Smardova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - F Pardy
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - M Doubek
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Y Brychtova
- Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - M Mraz
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - K Plevova
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - E Diviskova
- Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - A Oltova
- Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - J Mayer
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - S Pospisilova
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - M Trbusek
- 1] Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic [2] Department of Internal Medicine - Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
261
|
Sutton LA, Rosenquist R. Clonal evolution in chronic lymphocytic leukemia: impact of subclonality on disease progression. Expert Rev Hematol 2014; 8:71-8. [PMID: 25345442 DOI: 10.1586/17474086.2015.972930] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In recent years, next-generation sequencing has unraveled the molecular landscape in chronic lymphocytic leukemia with the discovery of a number of recurrently mutated genes. Mutations in several of these genes, such as NOTCH1, SF3B1 and BIRC3, are linked to a more aggressive disease with early disease progression, short time-to-first-treatment and even chemorefractoriness. Although in its infancy, we have also begun to understand the complex dynamics of subclonal diversity and its impact on disease outcome. From pioneering studies, we know that certain genetic events are found in the majority of chronic lymphocytic leukemia cells and are considered as 'clonal driver mutations' (e.g., +12, 13q-), whereas others, present only in a fraction of the tumor, are deemed to be 'subclonal driver mutations' for example, TP53 and SF3B1. Over the coming years, we need to gain a deeper insight into the dynamics of this subclonal architecture to understand how, at an individual level, chronic lymphocytic leukemia patients should be followed, which will be particularly relevant as novel targeted therapies begin to emerge.
Collapse
Affiliation(s)
- Lesley-Ann Sutton
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751-85, Uppsala, Sweden
| | | |
Collapse
|
262
|
Agnelli L, Neri A. Next-generation sequencing in multiple myeloma: insights into the molecular heterogeneity of the disease. Int J Hematol Oncol 2014. [DOI: 10.2217/ijh.14.30] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
SUMMARY Multiple myeloma (MM) is a still incurable malignant proliferation of clonal bone marrow plasma cells that is characterized by its variable clinical course, biology and molecular and genetic configuration. Given its relatively high incidence among hematological malignancies, a number of studies have taken advantage of large MM cohorts and used global gene, miRNA expression and genome-wide DNA profiling, and – more recently – next-generation sequencing (NGS) technology to investigate the genomic alterations underlying its bioclinical heterogeneity. Although still limited, NGS studies of MM have undoubtedly allowed a finer characterization of the molecular structure underlying the disease by further highlighting its heterogeneity and revealing novel molecular alterations. Herein, we present the main acquisitions on MM knowledge reached by the application of NGS.
Collapse
Affiliation(s)
- Luca Agnelli
- Department of Medical Sciences & Community Health, University of Milan, & UO Ematologia-CTMO, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonino Neri
- Department of Medical Sciences & Community Health, University of Milan, & UO Ematologia-CTMO, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
263
|
Gerrie AS, Huang SJT, Bruyere H, Dalal C, Hrynchak M, Karsan A, Ramadan KM, Smith AC, Tyson C, Toze CL, Gillan TL. Population-based characterization of the genetic landscape of chronic lymphocytic leukemia patients referred for cytogenetic testing in British Columbia, Canada: the role of provincial laboratory standardization. Cancer Genet 2014; 207:316-25. [PMID: 25441686 DOI: 10.1016/j.cancergen.2014.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 08/14/2014] [Accepted: 08/21/2014] [Indexed: 01/23/2023]
Abstract
Detection of recurrent chromosome abnormalities by fluorescence in situ hybridization (FISH) is an essential component of care in chronic lymphocytic leukemia (CLL) patients. In the province of British Columbia (BC), Canada, population 4.6 million, CLL patients receive uniform evaluation and therapy with FISH testing performed in three jurisdictions. The aims of this study were to (i) validate CLL-FISH testing among the BC cytogenetic laboratories to ensure standardization of results and (ii) characterize population-level CLL-FISH abnormalities by pooling provincial data. From 2004 to 2011, 585 consecutive patients underwent pretreatment CLL-FISH testing at laboratory A (50.1%), laboratory B (32.3%), or laboratory C (17.6%). For validation purposes, 26 CLL-FISH abnormalities were tested by each laboratory's protocol, with 91% result concordance. Discordant results involved percent abnormalities at or near cutoff values; therefore, a 10% universal cutoff was established when pooling results. Applying the universal cutoff to the provincial cohort, CLL-FISH abnormalities were detected in 74.9%: 54.9% 13q-, 18.8% +12, 8.5% 11q-, and 7.7% 17p-. In this large population-based cohort of patients referred for CLL-FISH testing, frequencies of abnormalities detected by FISH analysis were highly consistent with those reported in single-institution and clinical trial populations. Provinces or districts that work together to care for CLL patients can effectively pool data with appropriate laboratory validation to ensure standardization of results.
Collapse
Affiliation(s)
- Alina S Gerrie
- Leukemia/BMT Program of BC, Vancouver General Hospital and British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada
| | - Steven J T Huang
- Leukemia/BMT Program of BC, Vancouver General Hospital and British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada; Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Helene Bruyere
- Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Chinmay Dalal
- Leukemia/BMT Program of BC, Vancouver General Hospital and British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada
| | - Monica Hrynchak
- Cytogenetics Laboratory, Royal Columbian Hospital, New Westminster, Canada
| | - Aly Karsan
- Cancer Genetics Laboratory, Pathology and Laboratory Medicine, British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada
| | - Khaled M Ramadan
- Division of Hematology, St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| | - Adam C Smith
- Cancer Genetics Laboratory, Pathology and Laboratory Medicine, British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada; Instituto de Pesquisa Pelé Pequeno Princípe, Curitiba, Brazil
| | - Christine Tyson
- Cytogenetics Laboratory, Royal Columbian Hospital, New Westminster, Canada
| | - Cynthia L Toze
- Leukemia/BMT Program of BC, Vancouver General Hospital and British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada
| | - Tanya L Gillan
- Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
264
|
Shindiapina P, Brown JR, Danilov AV. A new hope: novel therapeutic approaches to treatment of chronic lymphocytic leukaemia with defects in TP53. Br J Haematol 2014; 167:149-61. [PMID: 25040077 DOI: 10.1111/bjh.13042] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chronic lymphocytic leukaemia (CLL) is an indolent B-cell malignancy with heterogeneous outcomes. Chromosomal abnormalities in CLL are predictive of the natural disease course; del(11q) and del(17p) are recognized as high risk genetic lesions. Del(17p) is associated with an impaired function of TP53, a key tumour suppressor, and is particularly problematic. Such patients respond poorly to chemo-immunotherapy and have significantly shorter survival compared to patients with standard and low-risk cytogenetics. While TP53 pathway defects are rare at initial diagnosis, their frequency increases in relapsed CLL. Until very recently, this group of patients represented an unmet clinical need with few therapeutic options. However, the advent of targeted therapies has expanded the drug armamentarium and introduced new hope for these highly refractory patients. Agents that target B-cell receptor signalling, BH3-mimetics and others induce apoptosis of the neoplastic B-cells in a TP53-independent manner. Their use in the clinic is associated with remarkable activity in patients with del(17p). In this review we discuss the frequency and clinical significance of del(17p) and genetic mutations leading to disrupted TP53, the putative role of other TP53 homologues, and the results of key clinical trials involving both conventional chemotherapy and novel agents.
Collapse
|
265
|
Affiliation(s)
- Robin Foà
- From the Department of Hematology, Sapienza University of Rome, Rome
| |
Collapse
|
266
|
Abstract
Over the past few years, massively parallel sequencing technologies have revealed with high resolution the tremendous genetic and epigenetic heterogeneity in chronic lymphocytic leukemia (CLL). We have learned how the molecular architecture differs not only between affected individuals but also within samples and over time. These insights have catalyzed our understanding of the pathobiology of CLL and point to critical signaling pathways in the development and progression of the disease. Several key driver alterations have been identified, which serve to refine prognostic schemata but also to inspire the development of new therapeutic strategies. Ongoing advances in technology promise to further elucidate the molecular basis of CLL, and this knowledge is anticipated to aid us in understanding and addressing the clinical challenge presented by the vast variability in the clinical course of patients with CLL.
Collapse
MESH Headings
- Chromosome Aberrations
- Epigenomics
- Genetic Heterogeneity
- Genome/genetics
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Michaela Gruber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Broad Institute, Cambridge, MA; Department of Internal Medicine I, Division of Haematology and Haemostaseology, Medical University of Vienna, Vienna, Austria
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Broad Institute, Cambridge, MA; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
267
|
BTK inhibitors in chronic lymphocytic leukemia: a glimpse to the future. Oncogene 2014; 34:2426-36. [PMID: 24954503 DOI: 10.1038/onc.2014.181] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 05/16/2014] [Accepted: 05/16/2014] [Indexed: 12/14/2022]
Abstract
The treatment of chronic lymphocytic leukemia (CLL) with inhibitors targeting B cell receptor signaling and other survival mechanisms holds great promise. Especially the early clinical success of Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase (BTK), has received widespread attention. In this review we will focus on the fundamental and clinical aspects of BTK inhibitors in CLL, with emphasis on Ibrutinib as the best studied of this class of drugs. Furthermore, we summarize recent laboratory as well as clinical findings relating to the first cases of Ibrutinib resistance. Finally, we address combination strategies with Ibrutinib, and attempt to extrapolate its current status to the near future in the clinic.
Collapse
|
268
|
Abstract
In this issue of Blood, Rossi et al provide further evidence for clonal evolution in chronic lymphocytic leukemia (CLL) and demonstrate the clinical importance of small TP53-deleted subclones detected at diagnosis in determining the natural history of the disease.
Collapse
|
269
|
Strati P, Keating MJ, O'Brien SM, Ferrajoli A, Burger J, Faderl S, Tambaro FP, Jain N, Wierda WG. Outcomes of first-line treatment for chronic lymphocytic leukemia with 17p deletion. Haematologica 2014; 99:1350-5. [PMID: 24859876 DOI: 10.3324/haematol.2014.104661] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Although uncommon in treatment-naive patients with chronic lymphocytic leukemia, deletion 17p is a high-risk disease characteristic. We analyzed and reported outcomes for 63 patients with deletion 17p chronic lymphocytic leukemia who received first-line therapy at our institution; at time of first treatment, 81% had unmutated immunoglobulin heavy chain variable gene and 58% had complex karyotype. Forty-nine patients (76%) received first-line fludarabine, cyclophosphamide, rituximab-based therapy, 6 (11%) received rituximab-based and 8 (13%) received lenalidomide-based treatment. Overall, the complete plus nodular partial remission rate was 33%; on multivariable model, higher complete plus nodular partial remission rate was observed in patients with less than 50% cells positive for deletion 17p, and a higher probability of achieving at least a partial remission was observed with fludarabine, cyclophosphamide, rituximab-based treatment. After a median follow up of 33 months (range 1-89 months), the estimated median progression-free survival was 14 months (95% confidence interval 10-18) and estimated median overall survival was 63 months (95% confidence interval 43-83). In multivariable analysis, factors independently associated with longer progression-free survival were response to treatment and absence of complex karyotype. Achievement of complete plus nodular partial remission rate and mutated immunoglobulin heavy chain variable gene were independently associated with longer overall survival in multivariable model. Complex karyotype was associated with increased risk for Richter's transformation. New first-line strategies and agents must aim at both improving response and maintaining remission in patients with deletion 17p, particularly in the presence of complex karyotype.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | - Michael J Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | - Susan M O'Brien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | - Alessandra Ferrajoli
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | - Jan Burger
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | - Stefan Faderl
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | | | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| | - William G Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center Houston, TX, USA
| |
Collapse
|
270
|
Genetic abnormalities in chronic lymphocytic leukemia: where we are and where we go. BIOMED RESEARCH INTERNATIONAL 2014; 2014:435983. [PMID: 24967369 PMCID: PMC4054680 DOI: 10.1155/2014/435983] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/22/2014] [Indexed: 12/17/2022]
Abstract
Chromosomal abnormalities in chronic lymphocytic leukemia (CLL) are detected in up to 80% of patients. Among them, deletions of 11q, 13q, 17p, and trisomy 12 have a known prognostic value and play an important role in CLL pathogenesis and evolution, determining patients outcome and therapeutic strategies. Standard methods used to identify these genomic aberrations include both conventional G-banding cytogenetics (CGC) and fluorescence in situ hybridization (FISH). Although FISH analyses have been implemented as the gold standard, CGC allows the identification of chromosomal translocations and complex karyotypes, the latest associated with poor outcome. Genomic arrays have a higher resolution that allows the detection of cryptic abnormalities, although these have not been fully implemented in routine laboratories. In the last years, next generation sequencing (NGS) methods have identified a wide range of gene mutations (e.g., TP53, NOTCH1, SF3B1, and BIRC3) which have improved our knowledge about CLL development, allowing us to refine both the prognostic subgroups and better therapeutic strategies. Clonal evolution has also recently arisen as a key point in CLL, integrating cytogenetic alterations and mutations in a dynamic model that improve our understanding about its clinical course and relapse.
Collapse
|