1
|
Kwok M, Stankovic T. Expanding the universe of T-PLL targets. Blood 2025; 145:2239-2241. [PMID: 40372742 DOI: 10.1182/blood.2025028353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Affiliation(s)
- Marwan Kwok
- University of Birmingham
- Dana-Farber Cancer Institute
| | | |
Collapse
|
2
|
Thorvaldsdottir B, Mansouri L, Sutton LA, Nadeu F, Meggendorfer M, Parker H, Brieghel C, Laidou S, Moia R, Rossi D, Kotaskova J, Delgado J, Rodríguez-Vicente AE, Benito R, Rigolin GM, Bonfiglio S, Scarfò L, Mattsson M, Davis Z, Baliakas P, Rapado I, Miras F, Martinez-Lopez J, de la Serna J, Hernández Rivas JM, Larráyoz MJ, Calasanz MJ, Smedby KE, Espinet B, Puiggros A, Bullinger L, Bosch F, Tazón-Vega B, Baran-Marszak F, Oscier D, Nguyen-Khac F, Zenz T, Terol MJ, Cuneo A, Hernández-Sánchez M, Pospisilova S, Gaidano G, Niemann CU, Campo E, Strefford JC, Ghia P, Stamatopoulos K, Rosenquist R. ATM aberrations in chronic lymphocytic leukemia: del(11q) rather than ATM mutations is an adverse-prognostic biomarker. Leukemia 2025:10.1038/s41375-025-02615-5. [PMID: 40275070 DOI: 10.1038/s41375-025-02615-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Despite the well-established adverse impact of del(11q) in chronic lymphocytic leukemia (CLL), the prognostic significance of somatic ATM mutations remains uncertain. We evaluated the effects of ATM aberrations (del(11q) and/or ATM mutations) on time-to-first-treatment (TTFT) in 3631 untreated patients with CLL, in the context of IGHV gene mutational status and mutations in nine CLL-related genes. ATM mutations were present in 246 cases (6.8%), frequently co-occurring with del(11q) (112/246 cases, 45.5%). ATM-mutated patients displayed a different spectrum of genetic abnormalities when comparing IGHV-mutated (M-CLL) and unmutated (U-CLL) cases: M-CLL was enriched for SF3B1 and NFKBIE mutations, whereas U-CLL showed mutual exclusivity with trisomy 12 and TP53 mutations. Isolated ATM mutations were rare, affecting 1.2% of Binet A patients and <1% of M-CLL cases. While univariable analysis revealed shorter TTFT for Binet A patients with any ATM aberration compared to ATM-wildtype, multivariable analysis identified only del(11q), trisomy 12, SF3B1, and EGR2 mutations as independent prognosticators of shorter TTFT among Binet A patients and within M-CLL and U-CLL subgroups. These findings highlight del(11q), and not ATM mutations, as a key biomarker of increased risk of early progression and need for therapy, particularly in otherwise indolent M-CLL, providing insights into risk-stratification and therapeutic decision-making.
Collapse
Affiliation(s)
- Birna Thorvaldsdottir
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Larry Mansouri
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lesley-Ann Sutton
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ferran Nadeu
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | | | - Helen Parker
- Cancer Genomics, School for Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Christian Brieghel
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Hematology, Danish Cancer Institute, Copenhagen, Denmark
| | - Stamatia Laidou
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece
| | - Riccardo Moia
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Davide Rossi
- Clinic of Hematology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Laboratory of Experimental Hematology, Institute of Oncology Research, Universita' della Svizzera Italiana, Bellinzona, Switzerland
| | - Jana Kotaskova
- Department of Internal Medicine, Hematology & Oncology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Julio Delgado
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana E Rodríguez-Vicente
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Rocío Benito
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - Gian Matteo Rigolin
- Hematology - Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | | | - Lydia Scarfò
- IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Mattias Mattsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Zadie Davis
- Molecular Pathology Department, University Hospitals Dorset, Bournemouth, UK
| | - Panagiotis Baliakas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Inmaculada Rapado
- Hospital Universitario 12 Octubre, Madrid, Spain
- Spanish National Cancer Research (CNIO), Madrid, Spain
| | - Fatima Miras
- Hospital Universitario 12 Octubre, Madrid, Spain
| | - Joaquín Martinez-Lopez
- Hospital Universitario 12 Octubre, Madrid, Spain
- Spanish National Cancer Research (CNIO), Madrid, Spain
| | - Javier de la Serna
- Hospital Universitario 12 Octubre, Madrid, Spain
- Spanish National Cancer Research (CNIO), Madrid, Spain
| | - Jesús María Hernández Rivas
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | - María José Larráyoz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Spain, IdiSNA, Navarra Institute for Health Research, 31008, Pamplona, Spain
| | - María José Calasanz
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, 31008 Pamplona, Spain, IdiSNA, Navarra Institute for Health Research, 31008, Pamplona, Spain
| | - Karin E Smedby
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Blanca Espinet
- Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar and Translational Research on Hematological Neoplasms Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Anna Puiggros
- Molecular Cytogenetics Laboratory, Pathology Department, Hospital del Mar and Translational Research on Hematological Neoplasms Group, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - Lars Bullinger
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Hum-boldt-Universität zu Berlin, Berlin, Germany
| | - Francesc Bosch
- Department of Hematology, Hospital Universitari Vall d'Hebron (HUVH), Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Bárbara Tazón-Vega
- Department of Hematology, Hospital Universitari Vall d'Hebron (HUVH), Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fanny Baran-Marszak
- Service d'hématologie biologique Hôpital Avicenne Assistance Publique des Hôpitaux de Paris Bobigny France, Bobigny, France
| | - David Oscier
- Molecular Pathology Department, University Hospitals Dorset, Bournemouth, UK
| | - Florence Nguyen-Khac
- Sorbonne Université, Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, APHP, Paris, France
| | - Thorsten Zenz
- Department of Oncology and Haematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Maria Jose Terol
- Department of Hematology, INCLIVA Research Insitute, University of Valencia, Valencia, Spain
| | - Antonio Cuneo
- Hematology - Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - María Hernández-Sánchez
- Cancer Research Center (IBMCC) CSIC-University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica (IBSAL), Salamanca, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, Madrid, Spain
| | - Sarka Pospisilova
- Department of Internal Medicine, Hematology & Oncology, University Hospital Brno, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Carsten U Niemann
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Hematology, Danish Cancer Institute, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Elias Campo
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Jonathan C Strefford
- Cancer Genomics, School for Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Paolo Ghia
- IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Kostas Stamatopoulos
- Centre for Research and Technology Hellas, Institute of Applied Biosciences, Thessaloniki, Greece
| | - Richard Rosenquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Clinical Genetics and Genomics, Karolinska University Hospital, Solna, Sweden.
| |
Collapse
|
3
|
Brown JR, Li J, Eichhorst BF, Lamanna N, O’Brien SM, Tam CS, Qiu L, Huang R, Shi Y, Idoine A, Salmi T, Cohen AC, Shadman M. Acquired mutations in patients with relapsed/refractory CLL who progressed in the ALPINE study. Blood Adv 2025; 9:1918-1926. [PMID: 39853273 PMCID: PMC12008693 DOI: 10.1182/bloodadvances.2024014206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/26/2025] Open
Abstract
ABSTRACT Some patients with chronic lymphocytic leukemia who develop progressive disease (PD) during covalent Bruton tyrosine kinase (BTK) inhibitor treatment acquire resistance mutations in BTK or PLCG2. Here, we report gene mutation data from paired baseline and PD peripheral blood samples from 52 patients (zanubrutinib, n = 24; ibrutinib, n = 28) who, at an early median follow-up of 25.7 months, progressed on zanubrutinib or ibrutinib treatment in ALPINE. No BTK mutations were observed at baseline; at PD, 8 patients (zanubrutinib, n = 5; ibrutinib, n = 3) acquired 17 BTK mutations, 82.4% (zanubrutinib, n = 11/14; ibrutinib, n = 3/3) at C481. Non-C481 mutations occurred in 12.5% (3/24) of zanubrutinib-treated patients (L528W: n = 2; cancer cell fraction [CCF] = 9.58% and 17.6%; A428D: n = 1; CCF = 37.03%). At baseline, 48 of 52 patients had ≥1 driver gene mutation(s), most frequently in NOTCH1 (n = 21), TP53 (n = 19), BRAF (n = 10), SF3B1 (n = 8), and ATM (n = 8). At PD, acquired mutations occurred in 1 zanubrutinib-treated patient (TP53, XPO1) and 5 ibrutinib-treated patients (TP53, n = 1 patient; SETD2, n = 1; SF3B1, n = 1; ASXL1, n = 2). Baseline driver gene mutations were not associated with development of BTK mutations, but patients with ≥2 baseline driver gene mutations were more likely to acquire BTK mutations at PD. The short treatment duration and a low BTK mutations incidence suggests that mechanisms other than BTK/PLCG2 mutations drive most early PD. This trial was registered at www.ClinicalTrials.gov as #NCT03734016.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Female
- Humans
- Male
- Middle Aged
- Adenine/analogs & derivatives
- Adenine/therapeutic use
- Adenine/analogs & derivatives
- Agammaglobulinaemia Tyrosine Kinase/genetics
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Disease Progression
- Drug Resistance, Neoplasm/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mutation
- Piperidines/therapeutic use
- Protein Kinase Inhibitors/therapeutic use
- Pyrazoles/therapeutic use
- Pyrimidines/therapeutic use
Collapse
Affiliation(s)
- Jennifer R. Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Barbara F. Eichhorst
- Department of Internal Medicine, University of Cologne, Center for Integrated Oncology Aachen Bonn Köln Düsseldorf, Cologne, Germany
| | - Nicole Lamanna
- Hematology/Oncology Division, Columbia University, New York, NY
| | - Susan M. O’Brien
- Division of Hematology/Oncology, University of California, Irvine, Irvine, CA
| | - Constantine S. Tam
- Department of Haematology, Alfred Hospital and Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Lugui Qiu
- National Clinical Research Center for Hematological Disorders, State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | | | | | | | - Tommi Salmi
- BeiGene International GmbH, Basel, Switzerland
| | | | - Mazyar Shadman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
4
|
Black GS, Huang X, Qiao Y, Moos P, Sampath D, Stephens DM, Woyach JA, Marth GT. Long-read single-cell RNA sequencing enables the study of cancer subclone-specific genotypes and phenotypes in chronic lymphocytic leukemia. Genome Res 2025; 35:686-697. [PMID: 39965935 PMCID: PMC12047255 DOI: 10.1101/gr.279049.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Bruton tyrosine kinase (BTK) inhibitors are effective for the treatment of chronic lymphocytic leukemia (CLL) due to BTK's role in B cell survival and proliferation. Treatment resistance is most commonly caused by the emergence of the hallmark BTK C481S mutation that inhibits drug binding. In this study, we aimed to investigate cancer subclones harboring a BTK C481S mutation and identify cells with co-occurring CLL driver mutations. In addition, we sought to determine whether BTK-mutated subclones exhibit distinct transcriptomic behavior when compared to other cancer subclones. To achieve these goals, we use scBayes, which integrates bulk DNA sequencing and single-cell RNA sequencing (scRNA-seq) data to genotype individual cells for subclone-defining mutations. Although the most common approach for scRNA-seq includes short-read sequencing, transcript coverage is limited due to the vast majority of the reads being concentrated at the priming end of the transcript. Here, we utilized MAS-seq, a long-read scRNA-seq technology, to substantially increase transcript coverage and expand the set of informative mutations to link cells to cancer subclones in six CLL patients who acquired BTK C481S mutations during BTK inhibitor treatment. In two patients who developed two independent BTK-mutated subclones, we find that most BTK-mutated cells have an additional CLL driver gene mutation. When examining subclone-specific gene expression, we find that in one patient, BTK-mutated subclones are transcriptionally distinct from the rest of the malignant B cell population with an overexpression of CLL-relevant genes.
Collapse
Affiliation(s)
- Gage S Black
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Xiaomeng Huang
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Yi Qiao
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Philip Moos
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, USA
| | - Deepa Sampath
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Deborah M Stephens
- Division of Hematology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Jennifer A Woyach
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Gabor T Marth
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA;
| |
Collapse
|
5
|
Kato M, Nishino J, Nagai M, Rokutan H, Narushima D, Ono H, Hasegawa T, Imoto S, Matsui S, Tsunoda T, Shibata T. Comprehensive analysis of prognosis markers with molecular features derived from pan-cancer whole-genome sequences. Hum Genomics 2025; 19:39. [PMID: 40221813 PMCID: PMC11993945 DOI: 10.1186/s40246-025-00744-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Cancer prognosis markers are useful for treatment decisions; however, the omics-level landscape is not well understood across multiple cancer types. Pan-Cancer Analysis of Whole Genomes (PCAWG) provides unprecedented access to various types of molecular data, ranging from typical DNA mutations and RNA expressions to more deeply analyzed or whole-genomic features, such as HLA haplotypes and structural variations. We analyzed the PCAWG data of 13 cancer types from 1,514 patients to identify prognosis markers belonging to 17 molecular features in the survival analysis based on the Cox and Lasso regression methods. We found that germline features including HLA haplotypes, neoantigens, and the number of structural variations were associated with overall survival; however, mutational signatures were not. Measuring a few markers provided a sufficient prognostic performance evaluated by c-index for each cancer type. DNA markers demonstrated better or comparable prognostic performance compared to RNA markers in some cancer types. "Universal" markers strongly associated with overall survival across cancer types were not identified. These findings will give insights into the clinical implementation of prognosis markers.
Collapse
Affiliation(s)
- Mamoru Kato
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan.
- CREST, JST, Tokyo, Japan.
| | - Jo Nishino
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
- CREST, JST, Tokyo, Japan
| | - Momoko Nagai
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
- CREST, JST, Tokyo, Japan
| | - Hirofumi Rokutan
- Division of Cancer Genomics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Daichi Narushima
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Hanako Ono
- Division of Bioinformatics, Research Institute, National Cancer Center Japan, Tokyo, Japan
| | - Takanori Hasegawa
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shigeyuki Matsui
- CREST, JST, Tokyo, Japan
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tatsuhiko Tsunoda
- CREST, JST, Tokyo, Japan
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo, Japan
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, Research Institute, National Cancer Center Japan, Tokyo, Japan
- Laboratory of Molecular Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
Rodríguez-Sánchez A, Quijada-Álamo M, Pérez-Carretero C, Herrero AB, Arroyo-Barea A, Dávila-Valls J, Rubio A, García de Coca A, Benito-Sánchez R, Rodríguez-Vicente AE, Hernández-Rivas JM, Hernández-Sánchez M. SAMHD1 dysfunction impairs DNA damage response and increases sensitivity to PARP inhibition in chronic lymphocytic leukemia. Sci Rep 2025; 15:10446. [PMID: 40140468 PMCID: PMC11947222 DOI: 10.1038/s41598-025-93629-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a clinically and genetically heterogenous disease. Recent next-generation sequencing (NGS) studies have uncovered numerous low-frequency mutated genes in CLL patients, with SAMHD1 emerging as a candidate driver gene. However, the biological and clinical implications of SAMHD1 mutations remain unclear. Using CRISPR/Cas9, we generated CLL models to investigate the impact of SAMHD1 deficiency on pathogenesis and explore therapeutic strategies. Moreover, we performed NGS in treatment-naïve CLL patients to characterize SAMHD1 mutations and employed RNA-sequencing to evaluate their clinical significance. Our study shows that SAMHD1 inactivation impairs the DNA damage response by reducing homologous recombination efficiency through BRCA1 and RAD51 dysregulation. Importantly, SAMHD1 colocalizes with BRCA1 at DNA damage sites in CLL cells. This research also identifies that SAMHD1-mutated cells are more sensitive to PARP inhibition. Clinically, SAMHD1 dysfunction negatively impacts clinical outcome of CLL cases: SAMHD1 mutations reduce failure-free survival (median 46 vs 57 months, p = 0.033), while low SAMHD1 expression associates with shorter time to first treatment (median 47 vs 77 months; p = 0.00073). Overall, this study elucidates that SAMHD1 dysfunction compromises DNA damage response mechanisms, potentially contributing to unfavorable clinical outcomes in CLL, and proposes PARP-inhibitors as a potential therapeutic approach for SAMHD1-mutated CLL cells.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- SAM Domain and HD Domain-Containing Protein 1/genetics
- SAM Domain and HD Domain-Containing Protein 1/metabolism
- DNA Damage
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
- Mutation
- Female
- Male
- BRCA1 Protein/metabolism
- BRCA1 Protein/genetics
- Middle Aged
- Aged
- CRISPR-Cas Systems
- Cell Line, Tumor
Collapse
Affiliation(s)
- Alberto Rodríguez-Sánchez
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Miguel Quijada-Álamo
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claudia Pérez-Carretero
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ana B Herrero
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Departamento de Medicina, Unidad de Medicina Molecular, Universidad de Salamanca, Salamanca, Spain
| | - Andrés Arroyo-Barea
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Julio Dávila-Valls
- Servicio de Hematología, Hospital Nuestra Señora de Sonsoles, SACYL, Ávila, Spain
| | - Araceli Rubio
- Servicio de Hematología, Hospital Miguel Servet, SERGAS, Zaragoza, Spain
| | | | - Rocío Benito-Sánchez
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Ana E Rodríguez-Vicente
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain
- Servicio de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - María Hernández-Sánchez
- Centro de Investigación del Cáncer, Universidad de Salamanca, IBSAL, IBMCC, CSIC, Salamanca, Spain.
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
7
|
Odintsov I, Papke DJ, George S, Padera RF, Hornick JL, Siegmund SE. Genomic Profiling of Cardiac Angiosarcoma Reveals Novel Targetable KDR Variants, Recurrent MED12 Mutations, and a High Burden of Germline POT1 Alterations. Clin Cancer Res 2025; 31:1091-1102. [PMID: 39820259 DOI: 10.1158/1078-0432.ccr-24-3277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/04/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Cardiac angiosarcoma is a rare, aggressive malignancy with limited treatment options. Both sporadic and familial cases occur, with recent links to germline POT1 mutations. The genomic landscape of this disease is poorly understood. EXPERIMENTAL DESIGN We conducted comprehensive genomic profiling of cardiac angiosarcoma to assess the burden of germline predisposition and identify other recurrent genomic alterations of clinical significance. RESULTS Six patients were female, and four were male. The median age at presentation was 40 years (range, 21-69 years). All cases with available follow-up exhibited an aggressive clinical course (6/8 patients died of disease). KDR alterations, including novel structural variants, were found in 9/11 cases at a rate significantly higher than that in noncardiac angiosarcomas. POT1 mutations were present in 45.5% of cardiac angiosarcoma cases. In three of five POT1-mutant cases, the germline status was confirmed through testing of normal tissue, and in one additional case, the germline status was inferred with high probability through allele frequency analysis. Additionally, we identified novel recurrent MED12 exon 2 mutations in POT1 wild-type cardiac angiosarcoma, suggesting an alternative path to cardiac angiosarcoma oncogenesis. CONCLUSIONS Cardiac angiosarcoma demonstrates a unique genetic profile, distinct from noncardiac angiosarcoma. This study highlights the role of germline POT1 burden on cardiac angiosarcoma development and demonstrates recurrent MED12 alterations for the first time. The reported KDR variants provide a potential avenue for the treatment of this aggressive disease. Given the prevalence of germline POT1 mutations reported in this study, germline genetic testing should be considered in patients diagnosed with cardiac angiosarcoma.
Collapse
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - David J Papke
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Suzanne George
- Sarcoma Division, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Stephanie E Siegmund
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
8
|
Arakelyan A, Sirunyan T, Khachatryan G, Hakobyan S, Minasyan A, Nikoghosyan M, Hakobyan M, Chavushyan A, Martirosyan G, Hakobyan Y, Binder H. Assigning Transcriptomic Subtypes to Chronic Lymphocytic Leukemia Samples Using Nanopore RNA-Sequencing and Self-Organizing Maps. Cancers (Basel) 2025; 17:964. [PMID: 40149301 PMCID: PMC11940626 DOI: 10.3390/cancers17060964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Massively parallel sequencing technologies have advanced chronic lymphocytic leukemia (CLL) diagnostics and precision oncology. Illumina platforms, while offering robust performance, require substantial infrastructure investment and a large number of samples for cost-efficiency. Conversely, third-generation long-read nanopore sequencing from Oxford Nanopore Technologies (ONT) can significantly reduce sequencing costs, making it a valuable tool in resource-limited settings. However, nanopore sequencing faces challenges with lower accuracy and throughput than Illumina platforms, necessitating additional computational strategies. In this paper, we demonstrate that integrating publicly available short-read data with in-house generated ONT data, along with the application of machine learning approaches, enables the characterization of the CLL transcriptome landscape, the identification of clinically relevant molecular subtypes, and the assignment of these subtypes to nanopore-sequenced samples. Methods: Public Illumina RNA sequencing data for 608 CLL samples were obtained from the CLL-Map Portal. CLL transcriptome analysis, gene module identification, and transcriptomic subtype classification were performed using the oposSOM R package for high-dimensional data visualization with self-organizing maps. Eight CLL patients were recruited from the Hematology Center After Prof. R. Yeolyan (Yerevan, Armenia). Sequencing libraries were prepared from blood total RNA using the PCR-cDNA sequencing-barcoding kit (SQK-PCB109) following the manufacturer's protocol and sequenced on an R9.4.1 flow cell for 24-48 h. Raw reads were converted to TPM values. These data were projected into the SOMs space using the supervised SOMs portrayal (supSOM) approach to predict the SOMs portrait of new samples using support vector machine regression. Results: The CLL transcriptomic landscape reveals disruptions in gene modules (spots) associated with T cell cytotoxicity, B and T cell activation, inflammation, cell cycle, DNA repair, proliferation, and splicing. A specific gene module contained genes associated with poor prognosis in CLL. Accordingly, CLL samples were classified into T-cell cytotoxic, immune, proliferative, splicing, and three mixed types: proliferative-immune, proliferative-splicing, and proliferative-immune-splicing. These transcriptomic subtypes were associated with survival orthogonal to gender and mutation status. Using supervised machine learning approaches, transcriptomic subtypes were assigned to patient samples sequenced with nanopore sequencing. Conclusions: This study demonstrates that the CLL transcriptome landscape can be parsed into functional modules, revealing distinct molecular subtypes based on proliferative and immune activity, with important implications for prognosis and treatment that are orthogonal to other molecular classifications. Additionally, the integration of nanopore sequencing with public datasets and machine learning offers a cost-effective approach to molecular subtyping and prognostic prediction, facilitating more accessible and personalized CLL care.
Collapse
Affiliation(s)
- Arsen Arakelyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
- Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia
| | - Tamara Sirunyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
| | - Gisane Khachatryan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
- Institute of Biomedicine and Pharmacy, Russian-Armenian University, Yerevan 0051, Armenia
| | - Siras Hakobyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
- Armenian Bioinformatics Institute, Yerevan 0014, Armenia;
| | - Arpine Minasyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
| | - Maria Nikoghosyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
- Armenian Bioinformatics Institute, Yerevan 0014, Armenia;
| | - Meline Hakobyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
| | | | - Gevorg Martirosyan
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia (S.H.); (M.N.)
| | - Yervand Hakobyan
- Hematology Center After Prof. R. Yeolyan MoH RA, Yerevan 0014, Armenia
| | - Hans Binder
- Armenian Bioinformatics Institute, Yerevan 0014, Armenia;
- Interdisciplinary Center for Bioinformatics, Leipzig University, 04109 Leipzig, Germany
| |
Collapse
|
9
|
Hallek M. Chronic Lymphocytic Leukemia: 2025 Update on the Epidemiology, Pathogenesis, Diagnosis, and Therapy. Am J Hematol 2025; 100:450-480. [PMID: 39871707 PMCID: PMC11803567 DOI: 10.1002/ajh.27546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 01/29/2025]
Abstract
DISEASE OVERVIEW Chronic lymphocytic leukemia (CLL) is the most frequent type of leukemia. It typically occurs in older patients and has a highly variable clinical course. Leukemic transformation is initiated by specific genomic alterations that interfere with the regulation of proliferation and apoptosis in clonal B-cells. DIAGNOSIS The diagnosis is established by blood counts, blood smears, and immunophenotyping of circulating B-lymphocytes, which identify a clonal B-cell population carrying the CD5 antigen as well as typical B-cell markers. PROGNOSIS AND STAGING Two clinical staging systems, Rai and Binet, provide prognostic information by using the results of physical examination and blood counts. Various biological and genetic markers provide additional prognostic information. Deletions of the short arm of chromosome 17 (del(17p)) and/or mutations of the TP53 gene predict a shorter time to progression with most targeted therapies. The CLL international prognostic index (CLL-IPI) integrates genetic, biological, and clinical variables to identify distinct risk groups of patients with CLL. The CLL-IPI retains its significance in the era of targeted agents, but the overall prognosis of CLL patients with high-risk stages has improved. THERAPY Only patients with active or symptomatic disease or with advanced Binet or Rai stages require therapy. When treatment is indicated, several therapeutic options exist: combinations of the BCL2 inhibitor venetoclax with obinutuzumab, or venetoclax with ibrutinib, or monotherapy with one of the inhibitors of Bruton tyrosine kinase (BTK). At relapse, the initial treatment may be repeated if the treatment-free interval exceeds 3 years. If the leukemia relapses earlier, therapy should be changed using an alternative regimen. FUTURE CHALLENGES Combinations of targeted agents now provide efficient therapies with a fixed duration that generate deep and durable remissions. These fixed-duration therapies have gained territory in the management of CLL, as they are cost-effective, avoid the emergence of resistance, and offer treatment free time to the patient. The cure rate of these novel combination regimens is unknown. Moreover, the optimal sequencing of targeted therapies remains to be determined. A medical challenge is to treat patients who are double-refractory to both BTK and BCL2 inhibitors. These patients need to be treated within experimental protocols using novel drugs.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Prognosis
- Mutation
Collapse
Affiliation(s)
- Michael Hallek
- Department I of Internal Medicine and Medical FacultyUniversity of CologneKölnGermany
- Center for Integrated Oncology Aachen Bonn Köln DüsseldorfKölnGermany
- Center of Excellence on “Cellular Stress Responses in Aging‐Associated Diseases,” University of CologneKölnGermany
- Center of Cancer Research Cologne EssenKölnGermany
- National Center for Tumor Diseases (NCT) WestKölnGermany
| |
Collapse
|
10
|
Ciantra Z, Paraskevopoulou V, Aifantis I. The rewired immune microenvironment in leukemia. Nat Immunol 2025; 26:351-365. [PMID: 40021898 DOI: 10.1038/s41590-025-02096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 01/15/2025] [Indexed: 03/03/2025]
Abstract
Leukemias are a class of human cancers that originate from hematopoietic progenitors and are characterized by extensive remodeling of the immune microenvironment. Leukemic cells, on transformation, acquire the ability to evade immune recognition but, despite undergoing genetic and epigenetic changes, retain their characteristic immature immune signature. For this and other reasons, leukemias are often refractory to immune therapies. In the present Review, we cover these areas as a means of improving outcomes from a deeper understanding of immune rewiring, inflammatory signaling and the barriers to successful implementation of immune therapies.
Collapse
Affiliation(s)
- Zoe Ciantra
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Varvara Paraskevopoulou
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Laura & Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Iannis Aifantis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
- Laura & Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Bonolo de Campos C, McCabe CE, Bruins LA, O'Brien DR, Brown S, Tschumper RC, Allmer C, Zhu YX, Rabe KG, Parikh SA, Kay NE, Yan H, Cerhan JR, Allan JN, Furman RR, Weinberg JB, Brander DM, Jelinek DF, Chesi M, Slager SL, Braggio E. Genomic characterization of chronic lymphocytic leukemia in patients of African ancestry. Blood Cancer J 2025; 15:14. [PMID: 39910036 PMCID: PMC11799526 DOI: 10.1038/s41408-024-01207-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 02/07/2025] Open
Abstract
Despite the considerable effort to characterize the genomic landscape of chronic lymphocytic leukemia (CLL), published data have been almost exclusively derived from patients of European Ancestry (EA), with significant underrepresentation of minorities, including patients of African Ancestry (AA). To begin to address this gap, we evaluated whether differences exist in the genetic and transcriptomic features of 157 AA and 440 EA individuals diagnosed with CLL. We sequenced 59 putative driver genes and found an increased frequency of high-impact mutations in AA CLL, including genes of the DNA damage repair (DDR) pathway. Telomere erosion was also increased in AA CLL, amplifying the notion of increased genomic instability in AA CLL. Furthermore, we found transcription enrichment of the Tumor Necrosis Factor-alpha (TNFα) Signaling via NF-κB pathway in AA CLL compared to EA CLL, suggesting that tumor promoting inflammation plays an important role in AA CLL. In summary, these results suggest that genomic instability and NF-kB activation is more prevalent in AA CLL than EA CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/ethnology
- Female
- Male
- Genomic Instability
- Middle Aged
- Aged
- Genomics/methods
- NF-kappa B/metabolism
- NF-kappa B/genetics
- Mutation
- Black People/genetics
- White People/genetics
- Aged, 80 and over
- Black or African American/genetics
Collapse
Affiliation(s)
| | - Chantal E McCabe
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Laura A Bruins
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Daniel R O'Brien
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - Sochilt Brown
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Cristine Allmer
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Yuan Xiao Zhu
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Kari G Rabe
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | | | - Neil E Kay
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Huihuang Yan
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - James R Cerhan
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - John N Allan
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Richard R Furman
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, USA
| | - J Brice Weinberg
- Divisions of Hematology and Hematologic Malignancies & Cellular Therapy & VA Medical Center, Durham, NC, USA
| | - Danielle M Brander
- Division of Hematologic Malignancy and Cellular Therapy, Duke Cancer Institute, Durham, NC, USA
| | | | - Marta Chesi
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Susan L Slager
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Esteban Braggio
- Division of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA.
| |
Collapse
|
12
|
Tosic N, Tomic Vujovic K, Vukovic V, Kotur N, Stankovic B, Marjanovic I, Antic D, Sarac S, Bibic T, Ivanovic J, Zukic B, Karan-Djurasevic T. High Expression Levels of the Long Non-Coding RNAs Lnc-IRF2-3 and Lnc-KIAA1755-4 Are Markers of Poor Prognosis in Chronic Lymphocytic Leukemia. Int J Mol Sci 2025; 26:1153. [PMID: 39940921 PMCID: PMC11817519 DOI: 10.3390/ijms26031153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) play complex roles at multiple levels of gene regulation, thus modulating key cellular processes involved in the pathogenesis and progression of cancer. Aberrant expression of lncRNAs has been reported in various malignancies, including chronic lymphocytic leukemia (CLL). We investigated the expression of lnc-IRF2-3 and lnc-KIAA1755-4 in peripheral blood mononuclear cells of 112 previously untreated CLL patients by quantitative reverse-transcriptase polymerase chain reaction. Both lncRNAs were found to be overexpressed in CLL samples in comparison to healthy controls, and their high levels were associated with adverse clinico-biological characteristics of patients at diagnosis. High lnc-IRF2-3 expression was associated with high leukocyte and lymphocyte counts, high β2-microglobulin, advanced Binet stage, unfavorable cytogenetics, CD38-positivity and IGHV-unmutated status. Regarding lnc-KIAA1755-4, its high expression was associated with high leukocyte count, lymphocyte count, β2-microglobulin, lactate dehydrogenase and low hemoglobin, as well as with IGHV-unmutated status. In addition, we observed shorter time to first treatment and overall survival of patients expressing high levels of both lncRNAs in comparison to low-expressing patients. In summary, our study showed that high lnc-IRF2-3 and lnc-KIAA1755-4 expression at diagnosis predicts poor survival in CLL. The mechanisms of their upregulation, as well as their specific targets in CLL cells, remain to be elucidated.
Collapse
MESH Headings
- Humans
- RNA, Long Noncoding/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Male
- Female
- Prognosis
- Middle Aged
- Aged
- Biomarkers, Tumor/genetics
- Gene Expression Regulation, Leukemic
- Aged, 80 and over
- Adult
- Leukocytes, Mononuclear/metabolism
- beta 2-Microglobulin
Collapse
Affiliation(s)
- Natasa Tosic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.T.); (N.K.); (B.S.); (I.M.); (B.Z.)
| | - Kristina Tomic Vujovic
- Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (K.T.V.); (V.V.); (D.A.); (S.S.); (T.B.); (J.I.)
| | - Vojin Vukovic
- Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (K.T.V.); (V.V.); (D.A.); (S.S.); (T.B.); (J.I.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nikola Kotur
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.T.); (N.K.); (B.S.); (I.M.); (B.Z.)
| | - Biljana Stankovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.T.); (N.K.); (B.S.); (I.M.); (B.Z.)
| | - Irena Marjanovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.T.); (N.K.); (B.S.); (I.M.); (B.Z.)
| | - Darko Antic
- Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (K.T.V.); (V.V.); (D.A.); (S.S.); (T.B.); (J.I.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Sofija Sarac
- Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (K.T.V.); (V.V.); (D.A.); (S.S.); (T.B.); (J.I.)
| | - Tamara Bibic
- Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (K.T.V.); (V.V.); (D.A.); (S.S.); (T.B.); (J.I.)
| | - Jelena Ivanovic
- Clinic for Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (K.T.V.); (V.V.); (D.A.); (S.S.); (T.B.); (J.I.)
| | - Branka Zukic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.T.); (N.K.); (B.S.); (I.M.); (B.Z.)
| | - Teodora Karan-Djurasevic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042 Belgrade, Serbia; (N.T.); (N.K.); (B.S.); (I.M.); (B.Z.)
| |
Collapse
|
13
|
Sobczyńska-Konefał A, Jasek M, Karabon L, Jaskuła E. Insights into genetic aberrations and signalling pathway interactions in chronic lymphocytic leukemia: from pathogenesis to treatment strategies. Biomark Res 2024; 12:162. [PMID: 39732734 DOI: 10.1186/s40364-024-00710-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024] Open
Abstract
Chronic lymphocytic leukemia (CLL) is prevalent in adults and is characterized by the accumulation of mature B cells in the blood, bone marrow, lymph nodes, and spleens. Recent progress in therapy and the introduction of targeted treatments [inhibitors of Bruton's tyrosine kinase (BTKi) or inhibitor of anti-apoptotic B-cell lymphoma-2 (Bcl-2i) protein (venetoclax)] in place of chemoimmunotherapy have significantly improved the outcomes of patients with CLL. These advancements have shifted the importance of traditional predictive markers, leading to a greater focus on resistance genes and reducing the significance of mutations, such as TP53 and del(17p). Despite the significant progress in CLL treatment, some patients still experience disease relapse. This is due to the substantial heterogeneity of CLL as well as the interconnected genetic resistance mechanisms and pathway adaptive resistance mechanisms to targeted therapies in CLL. Although the knowledge of the pathomechanism of CLL has expanded significantly in recent years, the precise origins of CLL and the interplay between various genetic factors remain incompletely understood, necessitating further research. This review enhances the molecular understanding of CLL by describing how BCR signalling, NF-κB PI3K/AKT, and ROR1 pathways sustain CLL cell survival, proliferation, and resistance to apoptosis. It also presents genetic and pathway-adaptive resistance mechanisms in CLL. Identifying B-cell receptor (BCR) signalling as a pivotal driver of CLL progression, the findings advocate personalized treatment strategies based on molecular profiling, emphasizing the need for further research to unravel the complex interplay between BCR signalling and its associated pathways to improve patient outcomes.
Collapse
Affiliation(s)
- Anna Sobczyńska-Konefał
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland
- Lower Silesian Oncology Hematology and Pulmonology Center, Ludwik Hirszfeld square 12, 53-413, Wroclaw, Poland
| | - Monika Jasek
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland
| | - Lidia Karabon
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland
| | - Emilia Jaskuła
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland.
- Lower Silesian Oncology Hematology and Pulmonology Center, Ludwik Hirszfeld square 12, 53-413, Wroclaw, Poland.
| |
Collapse
|
14
|
Charalampopoulou S, Chapiro E, Nadeu F, Zenz T, Beà S, Martínez-Farran A, Aymerich M, Rozman M, Roos-Weil D, Bernard O, Susin SA, Parker H, Walewska R, Oakes CC, Strefford JC, Campo E, Matutes E, Duran-Ferrer M, Nguyen-Khac F, Martín-Subero JI. Epigenetic features support the diagnosis of B-cell prolymphocytic leukemia and identify 2 clinicobiological subtypes. Blood Adv 2024; 8:6297-6307. [PMID: 39471431 DOI: 10.1182/bloodadvances.2024013327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 11/01/2024] Open
Abstract
ABSTRACT The recognition of B-cell prolymphocytic leukemia (B-PLL) as a separate entity is controversial based on the current classification systems. Here, we analyzed the DNA methylome of a cohort of 20 B-PLL cases diagnosed according to the guidelines of the International Consensus Classification/Fourth revised edition of the World Health Organization Classification, and compared them with chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), splenic marginal zone lymphoma (SMZL), and normal B-cell subpopulations. Unsupervised principal component analyses suggest that B-PLL is epigenetically distinct from CLL, MCL, and SMZL, which is further supported by robust differential methylation signatures in B-PLL. We also observe that B-PLL can be segregated into 2 epitypes with differential clinicobiological characteristics. B-PLL epitype 1 carries lower immunoglobulin heavy variable somatic hypermutation and a less profound germinal center-related DNA methylation imprint than epitype 2. Furthermore, epitype 1 is significantly enriched in mutations affecting MYC and SF3B1, and displays DNA hypomethylation and gene upregulation signatures enriched in MYC targets. Despite the low sample size, patients from epitype 1 have an inferior overall survival than those of epitype 2. This study provides relevant insights into the biology and differential diagnosis of B-PLL, and potentially identifies 2 subgroups with distinct biological and clinical features.
Collapse
MESH Headings
- Humans
- Epigenesis, Genetic
- DNA Methylation
- Leukemia, Prolymphocytic, B-Cell/diagnosis
- Leukemia, Prolymphocytic, B-Cell/genetics
- Male
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Female
- Lymphoma, Mantle-Cell/genetics
- Lymphoma, Mantle-Cell/diagnosis
- Lymphoma, Mantle-Cell/mortality
- Aged
Collapse
Affiliation(s)
| | - Elise Chapiro
- Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, INSERM UMRS 1138, Drug Resistance in Hematological Malignancies Team, Paris, France
| | - Ferran Nadeu
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital and University of Zürich, Zurich, Switzerland
| | - Sílvia Beà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Pathology Department, Hematopathology Section, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Fonaments Clinics, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | | | - Marta Aymerich
- Pathology Department, Hematopathology Section, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Maria Rozman
- Pathology Department, Hematopathology Section, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Damien Roos-Weil
- Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, INSERM UMRS 1138, Drug Resistance in Hematological Malignancies Team, Paris, France
| | | | - Santos A Susin
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, INSERM UMRS 1138, Drug Resistance in Hematological Malignancies Team, Paris, France
| | - Helen Parker
- Cancer Genomics, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Renata Walewska
- Department of Molecular Pathology, University Hospitals Dorset, Bournemouth, United Kingdom
| | | | - Jonathan C Strefford
- Cancer Genomics, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Elías Campo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Pathology Department, Hematopathology Section, Hospital Clínic de Barcelona, Barcelona, Spain
- Departament de Fonaments Clinics, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Estela Matutes
- Pathology Department, Hematopathology Section, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Martí Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Florence Nguyen-Khac
- Service d'Hématologie Biologique, Hôpital Pitié-Salpêtrière, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, INSERM UMRS 1138, Drug Resistance in Hematological Malignancies Team, Paris, France
| | - José I Martín-Subero
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Departament de Fonaments Clinics, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| |
Collapse
|
15
|
Vijenthira A, Volpe VO, Sekar A, Santos Azevedo R, Mikhaleva M, Gibson CJ, Martindale SP, Fardoun R, Tyekucheva S, Ren Y, Fernandes SM, Knisbacher BA, Hahn CK, Getz G, Wu CJ, Davids MS, Brown JR. Myeloid clonal hematopoiesis of indeterminate potential in patients with chronic lymphocytic leukemia. Blood Adv 2024; 8:5949-5956. [PMID: 39321421 PMCID: PMC11629178 DOI: 10.1182/bloodadvances.2024013414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/26/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024] Open
Abstract
ABSTRACT Clonal hematopoiesis of indeterminate potential (CHIP) in patients with chronic lymphocytic leukemia (CLL) has not been extensively characterized. The objective of this study was to describe the prevalence of myeloid CHIP (M-CHIP) in patients with CLL, and to determine its association with time to first treatment (TTFT) and overall survival (OS). We retrospectively analyzed data from patients participating in a prospective CLL database at the Dana-Farber Cancer Institute who had standard-of-care targeted 95-gene next-generation sequencing (NGS) performed. A schema was devised to classify mutations as M-CHIP related. M-CHIP was analyzed as a binary (present/absent) and categorical (≥2 vs 1 vs 0 mutations) predictor. We included 966 patients (median age at time of NGS, 65 years; 38% female). Overall, 747 (77%) patients had NGS performed before CLL treatment, whereas 219 (23%) had it performed after receiving treatment. Median follow-up time from NGS was 1.9 years. The prevalence of M-CHIP in untreated (12%) and treated (24%) patients with CLL was similar to that described in previous literature. M-CHIP prevalence appeared to increase with age in untreated patients, but appeared consistent across age in treated patients, suggesting that treatment (85% had prior chemotherapy) may have an impact on M-CHIP emergence even in younger patients. The presence of ≥2 M-CHIP mutations was associated with OS, even accounting for prior treatment and age, but was driven by a small subset of patients (n = 28). M-CHIP was not associated with TTFT. These findings support continued work into characterizing the effects of M-CHIP in patients with CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Female
- Aged
- Male
- Clonal Hematopoiesis
- Middle Aged
- Mutation
- High-Throughput Nucleotide Sequencing
- Retrospective Studies
- Aged, 80 and over
- Adult
- Prognosis
Collapse
Affiliation(s)
- Abi Vijenthira
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Virginia O. Volpe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Aswin Sekar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Mariia Mikhaleva
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Christopher J. Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | | | - Rayan Fardoun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Svitlana Tyekucheva
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Yue Ren
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Binyamin A. Knisbacher
- The Mina and Everard Goodman Faculty of Life Sciences, Ramat Gan, Israel
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Cynthia K. Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Gad Getz
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
- Krantz Family Center for Cancer Research and Dept. of Pathology, Massachusetts General Hospital, Boston, MA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jennifer R. Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
Hunter S, Ryland G, Pang JM, Ninkovic S, Dun K, Seymour JF, Blombery P. Chronic lymphocytic leukemia with MDM2 amplification as an alternative pathway to TP53 dysfunction. Leuk Lymphoma 2024; 65:2215-2218. [PMID: 39230950 DOI: 10.1080/10428194.2024.2398659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/19/2024] [Accepted: 08/24/2024] [Indexed: 09/05/2024]
Affiliation(s)
- Sally Hunter
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Georgina Ryland
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Jia-Min Pang
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Slavisa Ninkovic
- Victorian Cancer Cytogenetics Service, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Karen Dun
- Victorian Cancer Cytogenetics Service, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
- Department of Haematology, St. Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - John F Seymour
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia
| | - Piers Blombery
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
- Clinical Haematology, Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Melbourne, VIC, Australia
| |
Collapse
|
17
|
Smith CIE, Burger JA, Zain R. Estimating the Number of Polygenic Diseases Among Six Mutually Exclusive Entities of Non-Tumors and Cancer. Int J Mol Sci 2024; 25:11968. [PMID: 39596040 PMCID: PMC11593959 DOI: 10.3390/ijms252211968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
In the era of precision medicine with increasing amounts of sequenced cancer and non-cancer genomes of different ancestries, we here enumerate the resulting polygenic disease entities. Based on the cell number status, we first identified six fundamental types of polygenic illnesses, five of which are non-cancerous. Like complex, non-tumor disorders, neoplasms normally carry alterations in multiple genes, including in 'Drivers' and 'Passengers'. However, tumors also lack certain genetic alterations/epigenetic changes, recently named 'Goners', which are toxic for the neoplasm and potentially constitute therapeutic targets. Drivers are considered essential for malignant transformation, whereas environmental influences vary considerably among both types of polygenic diseases. For each form, hyper-rare disorders, defined as affecting <1/108 individuals, likely represent the largest number of disease entities. Loss of redundant tumor-suppressor genes exemplifies such a profoundly rare mutational event. For non-tumor, polygenic diseases, pathway-centered taxonomies seem preferable. This classification is not readily feasible in cancer, but the inclusion of Drivers and possibly also of epigenetic changes to the existing nomenclature might serve as initial steps in this direction. Based on the detailed genetic alterations, the number of polygenic diseases is essentially countless, but different forms of nosologies may be used to restrict the number.
Collapse
Affiliation(s)
- C. I. Edvard Smith
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Alfred Nobels Allé 8 Floor 8, SE-141 52 Huddinge, Sweden;
- Karolinska ATMP Center, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, SE-141 86 Huddinge, Sweden
| | - Jan A. Burger
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Rula Zain
- Department of Laboratory Medicine, Karolinska Institutet, ANA Futura, Alfred Nobels Allé 8 Floor 8, SE-141 52 Huddinge, Sweden;
- Karolinska ATMP Center, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
- Centre for Rare Diseases, Department of Clinical Genetics, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
18
|
Piñeyroa JA, López-Oreja I, Nadeu F, Martínez-Farran A, Aróstegui JI, López-Guerra M, Correa JG, Fabregat A, Villamor N, Monge-Escatín I, Albiol N, Costa D, Aymerich M, Beà S, Campo E, Delgado J, Colomer D, Mozas P. Association of Genomic Alterations with the Presence of Serum Monoclonal Proteins in Chronic Lymphocytic Leukemia. Cells 2024; 13:1839. [PMID: 39594588 PMCID: PMC11592641 DOI: 10.3390/cells13221839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
The presence of a monoclonal protein detected by serum immunofixation electrophoresis (sIFE) has been reported as an adverse prognostic factor in chronic lymphocytic leukemia (CLL). However, the genetic underpinning of this finding has not been studied. We retrospectively studied 97 CLL patients with simultaneous information on sIFE and genetic alterations detected by next-generation sequencing. sIFE was positive in 49 patients. The most common isotypes were IgG κ (27%) and bi/triclonal (25%). A +sIFE was associated with a higher number of mutated genes [median 2 (range 0-3) vs. 0 (range 0-2), p = 0.006], and a higher frequency of unmutated IGHV status (60 vs. 29%, p = 0.004). An IgM monoclonal protein was associated with TP53 mutations (36% in IgM +sIFE vs. 12% in non-IgM +sIFE or -sIFE, p = 0.04), and bi/triclonal proteins with NOTCH1 mutations (33% in bi/triclonal vs. 9% in monoclonal +sIFE or -sIFE, p = 0.04). These data suggest an association between a +sIFE and a higher mutational burden, and some monoclonal isotypes with specific mutations.
Collapse
Affiliation(s)
- Juan A. Piñeyroa
- Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain; (J.A.P.); (J.G.C.); (N.A.); (J.D.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Facultat de Medicina i Ciènces de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Irene López-Oreja
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Ferran Nadeu
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ares Martínez-Farran
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
| | - Juan Ignacio Aróstegui
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Department of Immunology, Hospital Clínic, 08036 Barcelona, Spain
| | - Mónica López-Guerra
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Juan Gonzalo Correa
- Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain; (J.A.P.); (J.G.C.); (N.A.); (J.D.)
| | - Aleix Fabregat
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Department of Biochemistry and Molecular Genetics, Hospital Clínic, 08036 Barcelona, Spain
| | - Neus Villamor
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | | | - Nil Albiol
- Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain; (J.A.P.); (J.G.C.); (N.A.); (J.D.)
| | - Dolors Costa
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Marta Aymerich
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Sílvia Beà
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Facultat de Medicina i Ciènces de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Elías Campo
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Facultat de Medicina i Ciènces de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Julio Delgado
- Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain; (J.A.P.); (J.G.C.); (N.A.); (J.D.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Facultat de Medicina i Ciènces de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Dolors Colomer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
- Facultat de Medicina i Ciènces de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Hematopathology Unit, Department of Pathology, Hospital Clínic, 08036 Barcelona, Spain
| | - Pablo Mozas
- Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain; (J.A.P.); (J.G.C.); (N.A.); (J.D.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (I.L.-O.); (F.N.); (A.M.-F.); (J.I.A.); (M.L.-G.); (A.F.); (N.V.); (D.C.); (M.A.); (S.B.); (E.C.)
| |
Collapse
|
19
|
Sekar A, Griffin R, Parikh SA, Genovese G, Robinson DP, Norman AD, Olson JE, Rabe KG, Hoel MS, Boddicker NJ, Hampel PJ, Kay NE, Cerhan JR, Braggio E, Hanson CA, Vachon CM, Shanafelt TD, Ebert BL, Slager SL. Mosaic chromosomal alterations (mCAs) in individuals with monoclonal B-cell lymphocytosis (MBL). Blood Cancer J 2024; 14:193. [PMID: 39505849 PMCID: PMC11541990 DOI: 10.1038/s41408-024-01175-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
MBL is a precursor condition to chronic lymphocytic leukemia (CLL), characterized by monoclonal B-cells in blood. Mosaic chromosomal alterations (mCAs) are a form of clonal hematopoiesis that include gains, losses, and copy-neutral loss-of-heterozygosity of large DNA segments. Both MBL and mCAs have been found to increase the risk of CLL and lymphoid malignancies, and the aim of our study was to investigate how mCAs relate to MBL, which is currently unknown. We analyzed genetic, flow cytometric, and hematologic data from 4632 individuals from the Mayo Clinic Biobank and CLL Database. MBL was detected using flow cytometry and classified as high-count (HC) or low-count (LC) MBL based on clone size. mCAs were detected primarily from whole blood DNA using sensitive SNP-array-based analyses. mCAs commonly altered in CLL (deletion of 6q, 11q, 13q, 17p, and trisomy 12) were specific (>99%) to individuals with MBL and CLL. HC-MBL and LC-MBL individuals were 881-fold and 8-fold, respectively, more likely to harbor CLL-associated mCAs than those without MBL. The cell fraction bearing these mCAs typically exceeded the B-cell fraction, suggesting their origin prior to the B-cell lineage. Integrating genetic and blood count data enabled detecting HC-MBL with high specificity in a biobank sample. These results quantify the contribution of mCAs to MBL and could enable large studies of HC-MBL without the need for flow cytometric screening.
Collapse
Affiliation(s)
- Aswin Sekar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Rosalie Griffin
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | | | - Giulio Genovese
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Dennis P Robinson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Aaron D Norman
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Janet E Olson
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Kari G Rabe
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN, USA
| | - Mingma S Hoel
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Paul J Hampel
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - James R Cerhan
- Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Esteban Braggio
- Department of Hematology/Oncology, Mayo Clinic, Phoenix, AZ, USA
| | - Curtis A Hanson
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | | | - Tait D Shanafelt
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Susan L Slager
- Division of Computational Biology, Mayo Clinic, Rochester, MN, USA.
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Huang YJ, Lim JQ, Hsu JS, Kuo MC, Wang PN, Kao HW, Wu JH, Chen CC, Tsai SF, Ong CK, Shih LY. Next-Generation Integrated Sequencing Identifies Poor Prognostic Factors in Patients with MYD88-Mutated Chronic Lymphocytic Leukemia in Taiwan. Pathobiology 2024; 92:77-89. [PMID: 39357512 PMCID: PMC11965870 DOI: 10.1159/000541709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
INTRODUCTION Chronic lymphocytic leukemia (CLL) is the most common type of leukemia in the Western countries and is very rare in Asia. METHODS Peripheral blood or bone marrow mononuclear cells obtained at initial diagnosis from 215 patients with CLL were analyzed by using next-generation sequencing to investigate the ethnic differences in genetic abnormalities. RESULTS Whole-genome sequencing and whole-exome sequencing analyses on 30 cases showed that 9 genes, including IGLL5, MYD88, TCHH, DSCAM, AXDND1, BICRA, KMT2D, MYT1L, and RBM43, were more frequently mutated in our Taiwanese cohort compared with those of the Western cohorts. IGLL5, MYD88, and KMT2D genes were further analyzed by targeted sequencing in another 185 CLL patients, unraveling frequencies of 29.3%, 20.9%, and 15.0%, respectively. The most frequent positional mutation of MYD88 was V217F (26/45, 57.8%), followed by L265P (9/45, 20.0%). MYD88 mutations were significantly associated with IGLL5 mutations (p = 0.0004), mutated IGHV (p < 0.0001) and 13q deletion (p = 0.0164). CLL patients with co-occurrence of MYD88 mutations with KMT2D or/and IGLL5 mutations were associated with a significantly inferior survival compared to those with MYD88 mutation alone (not reached vs. 131.8 months, p = 0.007). In multivariate analysis, MYD88 mutation without KMT2D or IGLL5 mutations was an independently favorable predictor. CONCLUSIONS IGLL5, MYD88, and KMT2D mutations were enriched in Taiwanese CLL, and co-occurrence of MYD88 mutations with KMT2D or/and IGLL5 mutations was associated with a poorer prognosis. INTRODUCTION Chronic lymphocytic leukemia (CLL) is the most common type of leukemia in the Western countries and is very rare in Asia. METHODS Peripheral blood or bone marrow mononuclear cells obtained at initial diagnosis from 215 patients with CLL were analyzed by using next-generation sequencing to investigate the ethnic differences in genetic abnormalities. RESULTS Whole-genome sequencing and whole-exome sequencing analyses on 30 cases showed that 9 genes, including IGLL5, MYD88, TCHH, DSCAM, AXDND1, BICRA, KMT2D, MYT1L, and RBM43, were more frequently mutated in our Taiwanese cohort compared with those of the Western cohorts. IGLL5, MYD88, and KMT2D genes were further analyzed by targeted sequencing in another 185 CLL patients, unraveling frequencies of 29.3%, 20.9%, and 15.0%, respectively. The most frequent positional mutation of MYD88 was V217F (26/45, 57.8%), followed by L265P (9/45, 20.0%). MYD88 mutations were significantly associated with IGLL5 mutations (p = 0.0004), mutated IGHV (p < 0.0001) and 13q deletion (p = 0.0164). CLL patients with co-occurrence of MYD88 mutations with KMT2D or/and IGLL5 mutations were associated with a significantly inferior survival compared to those with MYD88 mutation alone (not reached vs. 131.8 months, p = 0.007). In multivariate analysis, MYD88 mutation without KMT2D or IGLL5 mutations was an independently favorable predictor. CONCLUSIONS IGLL5, MYD88, and KMT2D mutations were enriched in Taiwanese CLL, and co-occurrence of MYD88 mutations with KMT2D or/and IGLL5 mutations was associated with a poorer prognosis.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Myeloid Differentiation Factor 88/genetics
- Taiwan
- Female
- Male
- Prognosis
- Mutation
- Middle Aged
- Aged
- High-Throughput Nucleotide Sequencing
- Adult
- Aged, 80 and over
Collapse
Affiliation(s)
- Ying-Jung Huang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jing Quan Lim
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- ONCO-ACP, Duke-NUS Medical School, Singapore, Singapore
| | - Jacob Shujui Hsu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Chung Kuo
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Nan Wang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Hsiao-Wen Kao
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jin-Hou Wu
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chiu-Chen Chen
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Shih-Feng Tsai
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Choon Kiat Ong
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Lee-Yung Shih
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
21
|
Aslan B, Manyam G, Iles LR, Tantawy SI, Desikan SP, Wierda WG, Gandhi V. Transcriptomic and proteomic differences in BTK-WT and BTK-mutated CLL and their changes during therapy with pirtobrutinib. Blood Adv 2024; 8:4487-4501. [PMID: 38968154 PMCID: PMC11395759 DOI: 10.1182/bloodadvances.2023012360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024] Open
Abstract
ABSTRACT Covalent Bruton tyrosine kinase inhibitors (cBTKis), which bind to the BTK C481 residue, are now primary therapeutics for chronic lymphocytic leukemia (CLL). Alterations at C481, primarily C481S, prevent cBTKi binding and lead to the emergence of resistant clones. Pirtobrutinib is a noncovalent BTKi that binds to both wild-type (WT) and C481S-mutated BTK and has shown efficacy in BTK-WT and -mutated CLL patient groups. To compare baseline clinical, transcriptomic, and proteomic characteristics and their changes during treatment in these 2 groups, we used 67 longitudinal peripheral blood samples obtained during the first 3 cycles of treatment with pirtobrutinib from 18 patients with CLL (11 BTK-mutated, 7 BTK-WT) enrolled in the BRUIN (pirtobrutinib in relapsed or refractory B-cell malignancies) trial. Eastern Cooperative Oncology Group performance status, age, and Rai stage were similar in both groups. At baseline, lymph nodes were larger in the BTK-mutated cohort. All patients achieved partial remission within 4 cycles of pirtobrutinib. Lactate dehydrogenase and β2-microglobulin levels decreased in both cohorts after 1 treatment cycle. Expression analysis demonstrated upregulation of 35 genes and downregulation of 6 in the BTK-mutated group. Gene set enrichment analysis revealed that the primary pathways enriched in BTK-mutated cells were involved in cell proliferation, metabolism, and stress response. Pathways associated with metabolism and proliferation were downregulated in both groups during pirtobrutinib treatment. Proteomic data corroborated transcriptomic findings. Our data identified inherent differences between BTK-mutated and -WT CLL and demonstrated molecular normalization of plasma and omics parameters with pirtobrutinib treatment in both groups.
Collapse
MESH Headings
- Humans
- Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors
- Agammaglobulinaemia Tyrosine Kinase/metabolism
- Agammaglobulinaemia Tyrosine Kinase/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Mutation
- Middle Aged
- Transcriptome
- Pyrimidines/therapeutic use
- Pyrimidines/pharmacology
- Proteomics/methods
- Female
- Male
- Aged
- Piperidines/therapeutic use
- Piperidines/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Proteome
- Adenine/analogs & derivatives
- Adenine/therapeutic use
- Pyrazoles/therapeutic use
- Pyrazoles/pharmacology
- Aged, 80 and over
Collapse
Affiliation(s)
- Burcu Aslan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ganiraju Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lakesla R. Iles
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shady I. Tantawy
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sai Prasad Desikan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - William G. Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Varsha Gandhi
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
22
|
Meriranta L, Sorri S, Huse K, Liu X, Spasevska I, Zafar S, Chowdhury I, Dufva O, Sahlberg E, Tandarić L, Karjalainen-Lindsberg ML, Hyytiäinen M, Varjosalo M, Myklebust JH, Leppä S. Disruption of KLHL6 Fuels Oncogenic Antigen Receptor Signaling in B-Cell Lymphoma. Blood Cancer Discov 2024; 5:331-352. [PMID: 38630892 PMCID: PMC11369598 DOI: 10.1158/2643-3230.bcd-23-0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/31/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Pathomechanisms that activate oncogenic B-cell receptor (BCR) signaling in diffuse large B-cell lymphoma (DLBCL) are largely unknown. Kelch-like family member 6 (KLHL6) encoding a substrate-adapter for Cullin-3-RING E3 ubiquitin ligase with poorly established targets is recurrently mutated in DLBCL. By applying high-throughput protein interactome screens and functional characterization, we discovered that KLHL6 regulates BCR by targeting its signaling subunits CD79A and CD79B. Loss of physiologic KLHL6 expression pattern was frequent among the MCD/C5-like activated B-cell DLBCLs and was associated with higher CD79B levels and dismal outcome. Mutations in the bric-a-brac tramtrack broad domain of KLHL6 disrupted its localization and heterodimerization and increased surface BCR levels and signaling, whereas Kelch domain mutants had the opposite effect. Malfunctions of KLHL6 mutants extended beyond proximal BCR signaling with distinct phenotypes from KLHL6 silencing. Collectively, our findings uncover how recurrent mutations in KLHL6 alter BCR signaling and induce actionable phenotypic characteristics in DLBCL. Significance: Oncogenic BCR signaling sustains DLBCL cells. We discovered that Cullin-3-RING E3 ubiquitin ligase substrate-adapter KLHL6 targets BCR heterodimer (CD79A/CD79B) for ubiquitin-mediated degradation. Recurrent somatic mutations in the KLHL6 gene cause corrupt BCR signaling by disrupting surface BCR homeostasis. Loss of KLHL6 expression and mutant-induced phenotypes associate with targetable disease characteristics in B-cell lymphoma. See related commentary by Leveille et al. See related commentary by Corcoran et al.
Collapse
MESH Headings
- Humans
- Signal Transduction
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- CD79 Antigens/genetics
- CD79 Antigens/metabolism
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Mutation
- Cell Line, Tumor
- Carrier Proteins
Collapse
Affiliation(s)
- Leo Meriranta
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| | - Selma Sorri
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| | - Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B-cell malignancies and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Xiaonan Liu
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Ivana Spasevska
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B-cell malignancies and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Sadia Zafar
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Iftekhar Chowdhury
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Olli Dufva
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| | - Eerika Sahlberg
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Luka Tandarić
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | | | - Marko Hyytiäinen
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - June H. Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B-cell malignancies and Precision Immunotherapy Alliance, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Sirpa Leppä
- Research Programs Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland.
| |
Collapse
|
23
|
Fernandez MM, Yu L, Jia Q, Wang X, Hart KL, Jia Z, Lin RJ, Wang L. Engineering Oncogenic Hotspot Mutations on SF3B1 via CRISPR-Directed PRECIS Mutagenesis. CANCER RESEARCH COMMUNICATIONS 2024; 4:2498-2513. [PMID: 39194178 PMCID: PMC11421219 DOI: 10.1158/2767-9764.crc-24-0145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
SF3B1 is the most recurrently mutated RNA splicing gene in cancer. However, research of its pathogenic role has been hindered by a lack of disease-relevant cell line models. Here, our study compared four genome engineering platforms to establish SF3B1 mutant cell lines: CRISPR-Cas9 editing, AAV homology-directed repair editing, base editing (ABEmax, ABE8e), and prime editing (PE2, PE3, PE5max). We showed that prime editing via PE5max achieved the most efficient SF3B1 K700E editing across a wide range of cell lines. Our approach was further refined by coupling prime editing with a fluorescent reporter that leverages a SF3B1 mutation-responsive synthetic intron to mark successfully edited cells. By applying this approach, called prime editing coupled intron-assisted selection (PRECIS), we introduced the K700E hotspot mutation into two chronic lymphocytic leukemia cell lines, HG-3 and MEC-1. We demonstrated that our PRECIS-engineered cells faithfully recapitulate known mutant SF3B1 phenotypes, including altered splicing, copy number variations, and cell-growth defect. Moreover, we discovered that the SF3B1 mutation can cause the loss of Y chromosome in chronic lymphocytic leukemia. Our results showcase that PRECIS is an efficient and generalizable method for engineering genetically faithful SF3B1 mutant models. Our approach provides new insights on the role of SF3B1 mutation in cancer and enables the generation of SF3B1 mutant cell lines in relevant cellular context. SIGNIFICANCE This study developed an approach that can reliably and efficiently engineer SF3B1 mutation into different cellular contexts, thereby revealing novel roles of SF3B1 mutation in driving aberrant splicing, clonal evolution, and genome instability.
Collapse
Affiliation(s)
- Mike M. Fernandez
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
| | - Lei Yu
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Qiong Jia
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Xuesong Wang
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Kevyn L. Hart
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
| | - Zhenyu Jia
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Ren-Jang Lin
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, California.
| | - Lili Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California.
| |
Collapse
|
24
|
Schwab K, Riege K, Coronel L, Stanko C, Förste S, Hoffmann S, Fischer M. p53 target ANKRA2 cooperates with RFX7 to regulate tumor suppressor genes. Cell Death Discov 2024; 10:376. [PMID: 39181888 PMCID: PMC11344851 DOI: 10.1038/s41420-024-02149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
The transcription factor regulatory factor X 7 (RFX7) has been identified as a tumor suppressor that is recurrently mutated in lymphoid cancers and appears to be dysregulated in many other cancers. RFX7 is activated by the well-known tumor suppressor p53 and regulates several other known tumor suppressor genes. However, what other factors regulate RFX7 and its target genes remains unclear. Here, reporter gene assays were used to identify that RFX7 regulates the tumor suppressor gene PDCD4 through direct interaction with its X-box promoter motif. We utilized mass spectrometry to identify factors that bind to DNA together with RFX7. In addition to RFX7, we also identified RFX5, RFXAP, RFXANK, and ANKRA2 that bind to the X-box motif in the PDCD4 promoter. We demonstrate that ANKRA2 is a bona fide direct p53 target gene. We used transcriptome analyses in two cell systems to identify genes regulated by ANKRA2, its sibling RFXANK, and RFX7. These results revealed that ANKRA2 functions as a critical cofactor of RFX7, whereas RFXANK regulates largely distinct gene sets.
Collapse
Affiliation(s)
- Katjana Schwab
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Konstantin Riege
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Luis Coronel
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Clara Stanko
- Klinik für Innere Medizin II, Jena University Hospital, Comprehensive Cancer Center Central Germany, Jena, Germany
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine Jena (CMB), Jena University Hospital, Jena, Germany
| | - Silke Förste
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Steve Hoffmann
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany.
| | - Martin Fischer
- Computational Biology Group, Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany.
| |
Collapse
|
25
|
Nguyen-Khac F, Baron M, Guièze R, Feugier P, Fayault A, Raynaud S, Troussard X, Droin N, Damm F, Smagghe L, Susin S, Leblond V, Dartigeas C, Van den Neste E, Leprêtre S, Bernard OA, Roos-Weil D. Prognostic impact of genetic abnormalities in 536 first-line chronic lymphocytic leukaemia patients without 17p deletion treated with chemoimmunotherapy in two prospective trials: Focus on IGHV-mutated subgroups (a FILO study). Br J Haematol 2024; 205:495-502. [PMID: 38654616 DOI: 10.1111/bjh.19459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024]
Abstract
The potential prognostic influence of genetic aberrations on chronic lymphocytic leukaemia (CLL) can vary based on various factors, such as the immunoglobulin heavy variable (IGHV) status. We conducted an integrative analysis on genetic abnormalities identified through cytogenetics and targeted next-generation sequencing in 536 CLL patients receiving first-line chemo(immuno)therapies (CIT) as part of two prospective trials. We evaluated the prognostic implications of the main abnormalities, with specific attention to their relative impact according to IGHV status. In the entire cohort, unmutated (UM)-IGHV, complex karyotype, del(11q) and ATM mutations correlated significantly with shorter progression-free survival (PFS). Focusing on the subset of mutated IGHV (M-IGHV) patients, univariate analysis showed that complex karyotype, del(11q), SF3B1 and SAMHD1 mutations were associated with significant lower PFS. The prognostic influence varied based on the patient's IGHV status, as these abnormalities did not affect outcomes in the UM-IGHV subgroup. TP53 mutations had no significant impact on outcomes in the M-IGHV subgroup. Our findings highlight the diverse prognostic influence of genetic aberrations depending on the IGHV status in symptomatic CLL patients receiving first-line CIT. The prognosis of gene mutations and cytogenetic abnormalities needs to be investigated with a compartmentalized methodology, taking into account the IGVH status of patients receiving first-line BTK and/or BCL2 inhibitors.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Female
- Humans
- Male
- Middle Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Chromosome Deletion
- Chromosomes, Human, Pair 17/genetics
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Variable Region/genetics
- Immunotherapy/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Mutation
- Prognosis
- Prospective Studies
Collapse
Affiliation(s)
- Florence Nguyen-Khac
- Unité de Cytogénétique, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Marine Baron
- Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Romain Guièze
- Hematology Department, Clermont-Ferrand University Hospital, Clermont Auvergne University, Clermont-Ferrand, France
| | - Pierre Feugier
- Department of Hematology, University Hospital of Nancy, Nancy, France
| | | | - Sophie Raynaud
- Laboratory of Hematology, University Hospital of Nice, Nice, France
| | | | - Nathalie Droin
- Inserm U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Frederik Damm
- Department of Hematology, Oncology, and Cancer Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Luce Smagghe
- Unité de Cytogénétique, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | - Santos Susin
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
| | - Véronique Leblond
- Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| | | | - Eric Van den Neste
- Department of Hematology, Cliniques Universitaires Université Catholique de Louvain Saint-Luc, Bruxelles, Belgium
| | - Stéphane Leprêtre
- Department of Clinical Hematology, Centre Henri Becquerel, Rouen, France
| | - Olivier A Bernard
- Inserm U1170, Université Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Damien Roos-Weil
- Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, Centre de Recherche Des Cordeliers, INSERM, Sorbonne Université, Université Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France
- Service d'Hématologie Clinique, Hôpital Pitié-Salpêtrière, APHP, Sorbonne Université, Paris, France
| |
Collapse
|
26
|
Vervoordeldonk MYL, Hengeveld PJ, Levin MD, Langerak AW. B cell receptor signaling proteins as biomarkers for progression of CLL requiring first-line therapy. Leuk Lymphoma 2024; 65:1031-1043. [PMID: 38619476 DOI: 10.1080/10428194.2024.2341151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
The molecular landscape of chronic lymphocytic leukemia (CLL) has been extensively characterized, and various potent prognostic biomarkers were discovered. The genetic composition of the B-cell receptor (BCR) immunoglobulin (IG) was shown to be especially powerful for discerning indolent from aggressive disease at diagnosis. Classification based on the IG heavy chain variable gene (IGHV) somatic hypermutation status is routinely applied. Additionally, BCR IGH stereotypy has been implicated to improve risk stratification, through characterization of subsets with consistent clinical profiles. Despite these advances, it remains challenging to predict when CLL progresses to requiring first-line therapy, thus emphasizing the need for further refinement of prognostic indicators. Signaling pathways downstream of the BCR are essential in CLL pathogenesis, and dysregulated components within these pathways impact disease progression. Considering not only genomics but the entirety of factors shaping BCR signaling activity, this review offers insights in the disease for better prognostic assessment of CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Signal Transduction
- Disease Progression
- Biomarkers, Tumor/genetics
- Prognosis
Collapse
Affiliation(s)
- Mischa Y L Vervoordeldonk
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Paul J Hengeveld
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Anton W Langerak
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
27
|
Pollard S, Chan B, Gerrie AS, Raymakers AJN, Regier DA. Assay-guided treatment sequencing in chronic lymphocytic leukemia (CLL): a cost-effectiveness analysis. Sci Rep 2024; 14:17294. [PMID: 39068214 PMCID: PMC11283533 DOI: 10.1038/s41598-024-68431-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
Costly targeted cancer treatments challenge publicly-funded healthcare systems seeking to align expected benefit with value for money. In 2021, The Canadian Agency for Drugs and Technologies in Health (CADTH) published a provisional funding algorithm for risk-based treatment of chronic lymphocytic leukemia (CLL). We estimate the cost-effectiveness of this algorithm against current standard of care. We constructed a probabilistic Markov model comparing next generation sequencing (NGS) assay-guided front-line treatment of acalabrutinib versus venetoclax with obinutuzumab to a comparator wherein patients initiate acalabrutinib. The primary outcome was the incremental cost-effectiveness ratio (ICER) per quality-adjusted life-year (QALY) gained. Analyses were conducted from the British Columbia healthcare system perspective, with outcomes discounted at 1.5%. Assay informed treatment for patients with CLL resulted in an incremental cost effectiveness ratio of $18,040 (95% CI $16,491-$19,501) per quality adjusted life-year (QALY) gained. The probability of the NGS guided treatment algorithm being cost effective was 80% at a willingness to pay threshold of $50,000 and a corresponding ICER of $18,040. Assay-guided treatment sequencing adds additional costs to healthcare but may be a cost-effective intervention for adult patients with CLL. Integration of real-world evidence would improve the validity and reliability of model estimated for decision-makers.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/economics
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Humans
- Cost-Benefit Analysis
- High-Throughput Nucleotide Sequencing/economics
- High-Throughput Nucleotide Sequencing/methods
- Quality-Adjusted Life Years
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Bridged Bicyclo Compounds, Heterocyclic/economics
- Sulfonamides/economics
- Sulfonamides/therapeutic use
- Benzamides/therapeutic use
- Benzamides/economics
- Antibodies, Monoclonal, Humanized/economics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Markov Chains
- Pyrazines/economics
- Pyrazines/therapeutic use
- Algorithms
- Cost-Effectiveness Analysis
Collapse
Affiliation(s)
- Samantha Pollard
- Cancer Control Research, BC Cancer, Vancouver, Canada
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Brandon Chan
- Cancer Control Research, BC Cancer, Vancouver, Canada
| | - Alina S Gerrie
- Division of Medical Oncology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- BC Cancer, Centre for Lymphoid Cancer, Vancouver, Canada
| | | | - Dean A Regier
- Cancer Control Research, BC Cancer, Vancouver, Canada.
- School of Population and Public Health, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
28
|
Stamatopoulos K, Pavlova S, Al‐Sawaf O, Chatzikonstantinou T, Karamanidou C, Gaidano G, Cymbalista F, Kater AP, Rawstron A, Scarfò L, Ghia P, Rosenquist R. Realizing precision medicine in chronic lymphocytic leukemia: Remaining challenges and potential opportunities. Hemasphere 2024; 8:e113. [PMID: 39035106 PMCID: PMC11260284 DOI: 10.1002/hem3.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/29/2024] [Accepted: 06/04/2024] [Indexed: 07/23/2024] Open
Abstract
Patients with chronic lymphocytic leukemia (CLL) exhibit diverse clinical outcomes. An expanding array of genetic tests is now employed to facilitate the identification of patients with high-risk disease and inform treatment decisions. These tests encompass molecular cytogenetic analysis, focusing on recurrent chromosomal alterations, particularly del(17p). Additionally, sequencing is utilized to identify TP53 mutations and to determine the somatic hypermutation status of the immunoglobulin heavy variable gene. Concurrently, a swift advancement of targeted treatment has led to the implementation of novel strategies for patients with CLL, including kinase and BCL2 inhibitors. This review explores both current and emerging diagnostic tests aimed at identifying high-risk patients who should benefit from targeted therapies. We outline existing treatment paradigms, emphasizing the importance of matching the right treatment to the right patient beyond genetic stratification, considering the crucial balance between safety and efficacy. We also take into consideration the practical and logistical issues when choosing a management strategy for each individual patient. Furthermore, we delve into the mechanisms underlying therapy resistance and stress the relevance of monitoring measurable residual disease to guide treatment decisions. Finally, we underscore the necessity of aggregating real-world data, adopting a global perspective, and ensuring patient engagement. Taken together, we argue that precision medicine is not the mere application of precision diagnostics and accessibility of precision therapies in CLL but encompasses various aspects of the patient journey (e.g., lifestyle exposures and comorbidities) and their preferences toward achieving true personalized medicine for patients with CLL.
Collapse
Affiliation(s)
- Kostas Stamatopoulos
- Centre for Research and Technology HellasInstitute of Applied BiosciencesThessalonikiGreece
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
| | - Sarka Pavlova
- Department of Internal Medicine, Hematology and Oncology, and Institute of Medical Genetics and GenomicsUniversity Hospital Brno and Medical Faculty, Masaryk UniversityBrnoCzech Republic
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Othman Al‐Sawaf
- Department I of Internal Medicine and German CLL Study Group, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)University of Cologne, Faculty of Medicine and University Hospital of CologneCologneGermany
- Francis Crick Institute LondonLondonUK
- Cancer Institute, University College LondonLondonUK
| | | | - Christina Karamanidou
- Centre for Research and Technology HellasInstitute of Applied BiosciencesThessalonikiGreece
| | - Gianluca Gaidano
- Division of Haematology, Department of Translational MedicineUniversity of Eastern PiedmontNovaraItaly
| | | | - Arnon P. Kater
- Department of Hematology, Cancer Center AmsterdamAmsterdam University Medical Centers, University of AmsterdamAmsterdamThe Netherlands
| | - Andy Rawstron
- Haematological Malignancy Diagnostic ServiceLeeds Teaching Hospitals TrustLeedsUK
| | - Lydia Scarfò
- Medical SchoolUniversità Vita Salute San RaffaeleMilanoItaly
- Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanoItaly
| | - Paolo Ghia
- Medical SchoolUniversità Vita Salute San RaffaeleMilanoItaly
- Strategic Research Program on CLLIRCCS Ospedale San RaffaeleMilanoItaly
| | - Richard Rosenquist
- Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Clinical GeneticsKarolinska University HospitalStockholmSweden
| |
Collapse
|
29
|
Bonato A, Chakraborty S, Bomben R, Canarutto G, Felician G, Martines C, Zucchetto A, Pozzo F, Vujovikj M, Polesel J, Chiarenza A, Del Principe MI, Del Poeta G, D'Arena G, Marasca R, Tafuri A, Laurenti L, Piazza S, Dimovski AJ, Gattei V, Efremov DG. NFKBIE mutations are selected by the tumor microenvironment and contribute to immune escape in chronic lymphocytic leukemia. Leukemia 2024; 38:1511-1521. [PMID: 38486128 PMCID: PMC11216988 DOI: 10.1038/s41375-024-02224-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Loss-of-function mutations in NFKBIE, which encodes for the NF-κB inhibitor IκBε, are frequent in chronic lymphocytic leukemia (CLL) and certain other B-cell malignancies and have been associated with accelerated disease progression and inferior responses to chemotherapy. Using in vitro and in vivo murine models and primary patient samples, we now show that NFKBIE-mutated CLL cells are selected by microenvironmental signals that activate the NF-κB pathway and induce alterations within the tumor microenvironment that can allow for immune escape, including expansion of CD8+ T-cells with an exhausted phenotype and increased PD-L1 expression on the malignant B-cells. Consistent with the latter observations, we find increased expression of exhaustion markers on T-cells from patients with NFKBIE-mutated CLL. In addition, we show that NFKBIE-mutated murine CLL cells display selective resistance to ibrutinib and report inferior outcomes to ibrutinib treatment in NFKBIE-mutated CLL patients. These findings suggest that NFKBIE mutations can contribute to CLL progression through multiple mechanisms, including a bidirectional crosstalk with the microenvironment and reduced sensitivity to BTK inhibitor treatment.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Adenine/analogs & derivatives
- Adenine/pharmacology
- CD8-Positive T-Lymphocytes/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Mutation
- NF-kappa B/metabolism
- Piperidines/pharmacology
- Piperidines/therapeutic use
- Pyrazoles/pharmacology
- Pyrazoles/therapeutic use
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Tumor Escape/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Alice Bonato
- Molecular Hematology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Supriya Chakraborty
- Molecular Hematology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Riccardo Bomben
- Clinical and Experimental Onco-Hematology Unit, IRCCS Centro Di Riferimento Oncologico, Aviano, Italy
| | - Giulia Canarutto
- Computational Biology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Giulia Felician
- Molecular Hematology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Claudio Martines
- Molecular Hematology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Antonella Zucchetto
- Clinical and Experimental Onco-Hematology Unit, IRCCS Centro Di Riferimento Oncologico, Aviano, Italy
| | - Federico Pozzo
- Clinical and Experimental Onco-Hematology Unit, IRCCS Centro Di Riferimento Oncologico, Aviano, Italy
| | - Marija Vujovikj
- Research Center for Genetic Engineering and Biotechnology, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| | - Jerry Polesel
- Clinical and Experimental Onco-Hematology Unit, IRCCS Centro Di Riferimento Oncologico, Aviano, Italy
| | | | | | - Giovanni Del Poeta
- Hematology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Giovanni D'Arena
- Hematology and Stem Cell Transplantation Unit, IRCCS Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, Italy
| | - Roberto Marasca
- Division of Hematology, University of Modena and Reggio Emilia, Modena, Italy
| | - Agostino Tafuri
- Division of Hematology, University Hospital Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Luca Laurenti
- Hematology Unit, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Silvano Piazza
- Computational Biology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Aleksandar J Dimovski
- Research Center for Genetic Engineering and Biotechnology, Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
- Macedonian Academy of Sciences and Arts, Skopje, North Macedonia
| | - Valter Gattei
- Clinical and Experimental Onco-Hematology Unit, IRCCS Centro Di Riferimento Oncologico, Aviano, Italy
| | - Dimitar G Efremov
- Molecular Hematology Unit, International Center for Genetic Engineering and Biotechnology, Trieste, Italy.
- Macedonian Academy of Sciences and Arts, Skopje, North Macedonia.
| |
Collapse
|
30
|
Rathgeber AC, Ludwig LS, Penter L. Single-cell genomics-based immune and disease monitoring in blood malignancies. Clin Hematol Int 2024; 6:62-84. [PMID: 38884110 PMCID: PMC11180218 DOI: 10.46989/001c.117961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 12/25/2023] [Indexed: 06/18/2024] Open
Abstract
Achieving long-term disease control using therapeutic immunomodulation is a long-standing concept with a strong tradition in blood malignancies. Besides allogeneic hematopoietic stem cell transplantation that continues to provide potentially curative treatment for otherwise challenging diagnoses, recent years have seen impressive progress in immunotherapies for leukemias and lymphomas with immune checkpoint blockade, bispecific monoclonal antibodies, and CAR T cell therapies. Despite their success, non-response, relapse, and immune toxicities remain frequent, thus prioritizing the elucidation of the underlying mechanisms and identifying predictive biomarkers. The increasing availability of single-cell genomic tools now provides a system's immunology view to resolve the molecular and cellular mechanisms of immunotherapies at unprecedented resolution. Here, we review recent studies that leverage these technological advancements for tracking immune responses, the emergence of immune resistance, and toxicities. As single-cell immune monitoring tools evolve and become more accessible, we expect their wide adoption for routine clinical applications to catalyze more precise therapeutic steering of personal immune responses.
Collapse
Affiliation(s)
- Anja C. Rathgeber
- Berlin Institute for Medical Systems BiologyMax Delbrück Center for Molecular Medicine
- Department of Hematology, Oncology, and TumorimmunologyCharité - Universitätsmedizin Berlin
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin
| | - Leif S. Ludwig
- Berlin Institute for Medical Systems BiologyMax Delbrück Center for Molecular Medicine
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin
| | - Livius Penter
- Department of Hematology, Oncology, and TumorimmunologyCharité - Universitätsmedizin Berlin
- BIH Biomedical Innovation AcademyBerlin Institute of Health at Charité - Universitätsmedizin Berlin
| |
Collapse
|
31
|
Bertilaccio MTS, Chen SS. Mouse models of chronic lymphocytic leukemia and Richter transformation: what we have learnt and what we are missing. Front Immunol 2024; 15:1376660. [PMID: 38903501 PMCID: PMC11186982 DOI: 10.3389/fimmu.2024.1376660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
Although the chronic lymphocytic leukemia (CLL) treatment landscape has changed dramatically, unmet clinical needs are emerging, as CLL in many patients does not respond, becomes resistant to treatment, relapses during treatment, or transforms into Richter. In the majority of cases, transformation evolves the original leukemia clone into a diffuse large B-cell lymphoma (DLBCL). Richter transformation (RT) represents a dreadful clinical challenge with limited therapeutic opportunities and scarce preclinical tools. CLL cells are well known to highly depend on survival signals provided by the tumor microenvironment (TME). These signals enhance the frequency of immunosuppressive cells with protumor function, including regulatory CD4+ T cells and tumor-associated macrophages. T cells, on the other hand, exhibit features of exhaustion and profound functional defects. Overall immune dysfunction and immunosuppression are common features of patients with CLL. The interaction between malignant cells and TME cells can occur during different phases of CLL development and transformation. A better understanding of in vivo CLL and RT biology and the availability of adequate mouse models that faithfully recapitulate the progression of CLL and RT within their microenvironments are "conditio sine qua non" to develop successful therapeutic strategies. In this review, we describe the xenograft and genetic-engineered mouse models of CLL and RT, how they helped to elucidate the pathophysiology of the disease progression and transformation, and how they have been and might be instrumental in developing innovative therapeutic approaches to finally eradicate these malignancies.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Animals
- Tumor Microenvironment/immunology
- Humans
- Mice
- Disease Models, Animal
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
Collapse
Affiliation(s)
| | - Shih-Shih Chen
- Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| |
Collapse
|
32
|
Govindasamy C, El Newehy AS, Hussein-Al-Ali SH, Arulselvan P, Bharathi M, Parthasarathy S. Investigation of antiproliferative efficacy and apoptosis induction in leukemia cancer cells using irinotecan-loaded liposome-embedded nanofibers constructed from chitosan. Int J Biol Macromol 2024; 270:132284. [PMID: 38734353 DOI: 10.1016/j.ijbiomac.2024.132284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024]
Abstract
Liposomes and nanofibers have been implemented as efficacious vehicles for delivering anticancer drugs. With this view, this study explores the antiproliferative efficacy and apoptosis induction in leukemia cancer cells utilizing irinotecan-loaded liposome-embedded nanofibers fabricated from chitosan, a biological source. Specifically, we investigate the effectiveness of poly(ε-caprolactone) (PCL)/chitosan (CS) (core)/irinotecan (CPT)nanofibers (termed PCL-CS10 CPT), PCL/chitosan/irinotecan (core)/PCL/chitosan (shell) nanofibers (termed CS/CPT/PCL/CS), and irinotecan-coloaded liposome-incorporated PCL/chitosan-chitosan nanofibers (termed CPT@Lipo/CS/PCL/CS) in releasing irinotecan in a controlled manner and treating leukemia cancer. The fabricated formulations were characterized utilizing Fourier transform infrared analysis, transmission electron microscopy, scanning electron microscopy, dynamic light scattering, zeta potential, and polydispersity index. Irinotecan was released in a controlled manner from nanofibers filled with liposomes over 30 days. The cell viability of the fabricated nanofibrous materials toward Human umbilical vein endothelial cells (HUVECs) non-cancerous cells after 168 h was >98 % ± 1 %. The CPT@Lipo/CS/PCL/CS nanofibers achieved maximal cytotoxicity of 85 % ± 2.5 % against K562 leukemia cancer cells. The CPT@Lipo/CS/PCL/CS NFs exhibit a three-stage drug release pattern and demonstrate significant in vitro cytotoxicity. These findings indicate the potential of these liposome-incorporated core-shell nanofibers for future cancer therapy.
Collapse
Affiliation(s)
- Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Ahmed S El Newehy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | | | - Palanisamy Arulselvan
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 602 105, India
| | - Muruganantham Bharathi
- Centre for Drug Discovery, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641021, India
| | - Surya Parthasarathy
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
33
|
Chen SS. Mouse models of CLL: In vivo modeling of disease initiation, progression, and transformation. Semin Hematol 2024; 61:201-207. [PMID: 38755077 DOI: 10.1053/j.seminhematol.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 05/18/2024]
Abstract
Chronic lymphocytic leukemia (CLL) is a highly complex disease characterized by the proliferation of CD5+ B cells in lymphoid tissues. Current modern treatments have brought significant clinical benefits to CLL patients. However, there are still unmet needs. Patients relapse on Bruton's tyrosine kinase inhibitors and BCL2 inhibitors and often develop more aggressive diseases including Richter transformation (RT), an incurable complication of up to ∼10% patients. This evidence underscores the need for improved immunotherapies, combination treatment strategies, and predictive biomarkers. A mouse model that can recapitulate human CLL disease and certain components of the tumor immune microenvironment represents a promising preclinical tool for such purposes. In this review, we provide an overview of CRISPR-engineered and xenograft mouse models utilizing either cell lines, or primary CLL cells suitable for studies of key events driving the disease onset, progression and transformation of CLL. We also review how CRISPR/Cas9 established mouse models carrying loss-of-function lesions allow one to study key mutations driving disease progression. Finally, we discuss how next generation humanized mice might improve to generation of faithful xenograft mouse models of human CLL.
Collapse
MESH Headings
- Animals
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Humans
- Mice
- Disease Models, Animal
- Disease Progression
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Cell Transformation, Neoplastic/metabolism
- Tumor Microenvironment/immunology
- CRISPR-Cas Systems
Collapse
Affiliation(s)
- Shih-Shih Chen
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York.
| |
Collapse
|
34
|
Maiques-Diaz A, Martin-Subero JI. Biological, prognostic, and therapeutic impact of the epigenome in CLL. Semin Hematol 2024; 61:172-180. [PMID: 38151379 DOI: 10.1053/j.seminhematol.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/22/2023] [Indexed: 12/29/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by widespread alterations in the genetic and epigenetic landscapes which seem to underlie the variable clinical manifestations observed in patients. Over the last decade, epigenomic studies have described the whole-genome maps of DNA methylation and chromatin features of CLL and normal B cells, identifying distinct epigenetic mechanisms operating in tumoral cells. DNA methylation analyses have identified that the CLL methylome contains imprints of the cell of origin, as well as of the proliferative history of the tumor cells, with both being strong independent prognostic predictors. Moreover, single-cell analysis revealed a higher degree of DNA methylation noise in CLL cells, which associates with transcriptional plasticity and disease aggressiveness. Integrative analysis of chromatin has uncovered chromatin signatures, as well as regulatory regions specifically active in each CLL subtype or in Richter transformed samples. Unique transcription factor (TF) binding motifs are overrepresented on those regions, suggesting that altered TF networks operate from disease initiation to progression as nongenetic factors mediating the oncogenic transcriptional profiles. Multiomics analysis has identified that response to treatment is modulated by an epigenetic imprint, and that treatments affect chromatin through the activity of particular set of TFs. Additionally, the epigenome is an axis of therapeutic vulnerability in CLL, as it can be targeted by inhibitors of histone modifying enzymes, that have shown promising preclinical results. Altogether, this review aims at summarizing the major findings derived from published literature to distill how altered epigenomic mechanisms contribute to CLL origin, evolution, clinical behavior, and response to treatment.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Prognosis
- Epigenesis, Genetic
- Epigenome
- DNA Methylation/genetics
- Epigenomics
- Chromatin/genetics
- Chromatin/metabolism
Collapse
Affiliation(s)
- Alba Maiques-Diaz
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain
| | - Jose Ignacio Martin-Subero
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Departamento de Fundamentos Clínicos, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
35
|
Kwok M, Agathanggelou A, Stankovic T. DNA damage response defects in hematologic malignancies: mechanistic insights and therapeutic strategies. Blood 2024; 143:2123-2144. [PMID: 38457665 DOI: 10.1182/blood.2023019963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 02/15/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024] Open
Abstract
ABSTRACT The DNA damage response (DDR) encompasses the detection and repair of DNA lesions and is fundamental to the maintenance of genome integrity. Germ line DDR alterations underlie hereditary chromosome instability syndromes by promoting the acquisition of pathogenic structural variants in hematopoietic cells, resulting in increased predisposition to hematologic malignancies. Also frequent in hematologic malignancies are somatic mutations of DDR genes, typically arising from replication stress triggered by oncogene activation or deregulated tumor proliferation that provides a selective pressure for DDR loss. These defects impair homology-directed DNA repair or replication stress response, leading to an excessive reliance on error-prone DNA repair mechanisms that results in genomic instability and tumor progression. In hematologic malignancies, loss-of-function DDR alterations confer clonal growth advantage and adverse prognostic impact but may also provide therapeutic opportunities. Selective targeting of functional dependencies arising from these defects could achieve synthetic lethality, a therapeutic concept exemplified by inhibition of poly-(adenosine 5'-diphosphate ribose) polymerase or the ataxia telangiectasia and Rad 3 related-CHK1-WEE1 axis in malignancies harboring the BRCAness phenotype or genetic defects that increase replication stress. Furthermore, the role of DDR defects as a source of tumor immunogenicity, as well as their impact on the cross talk between DDR, inflammation, and tumor immunity are increasingly recognized, thus providing rationale for combining DDR modulation with immune modulation. The nature of the DDR-immune interface and the cellular vulnerabilities conferred by DDR defects may nonetheless be disease-specific and remain incompletely understood in many hematologic malignancies. Their comprehensive elucidation will be critical for optimizing therapeutic strategies to target DDR defects in these diseases.
Collapse
Affiliation(s)
- Marwan Kwok
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Angelo Agathanggelou
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Tatjana Stankovic
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
36
|
Kleinstern G, Boddicker NJ, O’Brien DR, Allmer C, Rabe KG, Norman AD, Griffin R, Yan H, Ma T, Call TG, Bruins L, Brown S, Bonolo de Campos C, Hanson CA, Leis JF, Ding W, Vachon CM, Kay NE, Oakes CC, Parker AS, Brander DM, Weinberg JB, Furman RR, Shanafelt TD, Cerhan JR, Parikh SA, Braggio E, Slager SL. Tumor mutational load is prognostic for progression to therapy among high-count monoclonal B-cell lymphocytosis. Blood Adv 2024; 8:2118-2129. [PMID: 38359367 PMCID: PMC11059316 DOI: 10.1182/bloodadvances.2023012242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/21/2023] [Accepted: 01/22/2024] [Indexed: 02/17/2024] Open
Abstract
ABSTRACT High-count monoclonal B-cell lymphocytosis (HCMBL) is a precursor condition to chronic lymphocytic leukemia (CLL). We have shown that among individuals with HCMBL, the CLL-International Prognostic Index (CLL-IPI) is prognostic for time-to-first therapy (TTFT). Little is known about the prognostic impact of somatically mutated genes among individuals with HCMBL. We sequenced DNA from 371 individuals with HCMBL using a targeted sequencing panel of 59 recurrently mutated genes in CLL to identify high-impact mutations. We compared the sequencing results with that of our treatment-naïve CLL cohort (N = 855) and used Cox regression to estimate hazard ratios and 95% confidence intervals (CIs) for associations with TTFT. The frequencies of any mutated genes were lower in HCMBL (52%) than CLL (70%). At 10 years, 37% of individuals with HCMBL with any mutated gene had progressed requiring treatment compared with 10% among individuals with HCMBL with no mutations; this led to 5.4-fold shorter TTFT (95% CI, 2.6-11.0) among HCMBL with any mutated gene vs none, independent of CLL-IPI. When considering individuals with low risk of progression according to CLL-IPI, those with HCMBL with any mutations had 4.3-fold shorter TTFT (95% CI, 1.6-11.8) vs those with none. Finally, when considering both CLL-IPI and any mutated gene status, we observed individuals with HCMBL who were high risk for both prognostic factors had worse prognosis than patients with low-risk CLL (ie, 5-year progression rate of 32% vs 21%, respectively). Among HCMBL, the frequency of somatically mutated genes at diagnosis is lower than that of CLL. Accounting for both the number of mutated genes and CLL-IPI can identify individuals with HCMBL with more aggressive clinical course.
Collapse
MESH Headings
- Humans
- Lymphocytosis/genetics
- Lymphocytosis/diagnosis
- Lymphocytosis/therapy
- Prognosis
- Mutation
- Male
- Female
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Middle Aged
- Aged
- Disease Progression
- B-Lymphocytes/metabolism
- B-Lymphocytes/pathology
- Adult
- Aged, 80 and over
- Lymphocyte Count
Collapse
Affiliation(s)
- Geffen Kleinstern
- School of Public Health, University of Haifa, Haifa, Israel
- Division of Computational Biology, Mayo Clinic, Rochester, MN
| | | | | | - Cristine Allmer
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | - Kari G. Rabe
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | | | - Rosalie Griffin
- Division of Computational Biology, Mayo Clinic, Rochester, MN
| | - Huihuang Yan
- Division of Computational Biology, Mayo Clinic, Rochester, MN
| | - Tao Ma
- Division of Computational Biology, Mayo Clinic, Rochester, MN
| | | | - Laura Bruins
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | - Sochilt Brown
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | | | - Curtis A. Hanson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Jose F. Leis
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | - Wei Ding
- Division of Hematology, Mayo Clinic, Rochester, MN
| | | | - Neil E. Kay
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Christopher C. Oakes
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus OH
- The Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH
| | | | | | - J. Brice Weinberg
- Department of Medicine, Duke University, Duke Cancer Institute, Durham, NC
- Department of Immunology, Duke University Medical Center, Durham, NC
- Durham Veterans Affairs Medical Center, Durham, NC
| | - Richard R. Furman
- Weill Cornell Medical College/New York Presbyterian Hospital, New York, NY
| | - Tait D. Shanafelt
- Department of Medicine, Division of Hematology, Stanford University, Stanford, CA
| | | | | | - Esteban Braggio
- Department of Hematology and Oncology, Mayo Clinic, Phoenix, AZ
| | - Susan L. Slager
- Division of Computational Biology, Mayo Clinic, Rochester, MN
- Division of Hematology, Mayo Clinic, Rochester, MN
| |
Collapse
|
37
|
Fernandez Botana I, Gonder S, Klapp V, Moussay E, Paggetti J. Eμ-TCL1 adoptive transfer mouse model of chronic lymphocytic leukemia. Methods Cell Biol 2024; 188:109-129. [PMID: 38880520 DOI: 10.1016/bs.mcb.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Despite being the most common adult leukemia in the western world, Chronic Lymphocytic Leukemia (CLL) remains a life-threatening and incurable disease. Efforts to develop new treatments are highly dependent on the availability of appropriate mouse models for pre-clinical testing. The Eμ-TCL1 mouse model is the most established pre-clinical approach to study CLL pathobiology and response to treatment, backed by numerous studies highlighting its resemblance to the most aggressive form of this malignancy. In contrast to the transgenic Eμ-TCL1 model, employing the adoptive transfer of Eμ-TCL1-derived splenocytes in immunocompetent C57BL/6 mice results in a comparably rapid (e.g., leukemic development within weeks compared to months in the transgenic model) and reliable model mimicking CLL. In this chapter, we would like to provide readers with a thoroughly optimized, detailed, and comprehensive protocol to use the adoptive transfer Eμ-TCL1 model in their research.
Collapse
Affiliation(s)
- Iria Fernandez Botana
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Susanne Gonder
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Vanessa Klapp
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Etienne Moussay
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg.
| | - Jerome Paggetti
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg.
| |
Collapse
|
38
|
Sciaccotta R, Gangemi S, Penna G, Giordano L, Pioggia G, Allegra A. Potential New Therapies "ROS-Based" in CLL: An Innovative Paradigm in the Induction of Tumor Cell Apoptosis. Antioxidants (Basel) 2024; 13:475. [PMID: 38671922 PMCID: PMC11047475 DOI: 10.3390/antiox13040475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chronic lymphocytic leukemia, in spite of recent advancements, is still an incurable disease; the majority of patients eventually acquire resistance to treatment through relapses. In all subtypes of chronic lymphocytic leukemia, the disruption of normal B-cell homeostasis is thought to be mostly caused by the absence of apoptosis. Consequently, apoptosis induction is crucial to the management of this illness. Damaged biological components can accumulate as a result of the oxidation of intracellular lipids, proteins, and DNA by reactive oxygen species. It is possible that cancer cells are more susceptible to apoptosis because of their increased production of reactive oxygen species. An excess of reactive oxygen species can lead to oxidative stress, which can harm biological elements like DNA and trigger apoptotic pathways that cause planned cell death. In order to upset the balance of oxidative stress in cells, recent therapeutic treatments in chronic lymphocytic leukemia have focused on either producing reactive oxygen species or inhibiting it. Examples include targets created in the field of nanomedicine, natural extracts and nutraceuticals, tailored therapy using biomarkers, and metabolic targets. Current developments in the complex connection between apoptosis, particularly ferroptosis and its involvement in epigenomics and alterations, have created a new paradigm.
Collapse
Affiliation(s)
- Raffaele Sciaccotta
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Giuseppa Penna
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Laura Giordano
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 98164 Messina, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (R.S.); (G.P.); (L.G.)
| |
Collapse
|
39
|
Sud A, Parry EM, Wu CJ. The molecular map of CLL and Richter's syndrome. Semin Hematol 2024; 61:73-82. [PMID: 38368146 PMCID: PMC11653080 DOI: 10.1053/j.seminhematol.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 02/19/2024]
Abstract
Clonal expansion of B-cells, from the early stages of monoclonal B-cell lymphocytosis through to chronic lymphocytic leukemia (CLL), and then in some cases to Richter's syndrome (RS) provides a comprehensive model of cancer evolution, notable for the marked morphological transformation and distinct clinical phenotypes. High-throughput sequencing of large cohorts of patients and single-cell studies have generated a molecular map of CLL and more recently, of RS, yielding fundamental insights into these diseases and of clonal evolution. A selection of CLL driver genes have been functionally interrogated to yield novel insights into the biology of CLL. Such findings have the potential to impact patient care through risk stratification, treatment selection and drug discovery. However, this molecular map remains incomplete, with extant questions concerning the origin of the B-cell clone, the role of the TME, inter- and intra-compartmental heterogeneity and of therapeutic resistance mechanisms. Through the application of multi-modal single-cell technologies across tissues, disease states and clinical contexts, these questions can now be addressed with the answers holding great promise of generating translatable knowledge to improve patient care.
Collapse
Affiliation(s)
- Amit Sud
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA; Department of Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Erin M Parry
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA.
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA; Department of Medicine, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
40
|
Tausch E, López C, Stilgenbauer S, Siebert R. Genetic alterations in chronic lymphocytic leukemia and plasma cell neoplasms - a practical guide to WHO HAEM5. MED GENET-BERLIN 2024; 36:47-57. [PMID: 38835970 PMCID: PMC11006374 DOI: 10.1515/medgen-2024-2006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours (WHO-HAEM5) provides a revised classification of lymphoid malignancies including chronic lymphocytic leukemia (CLL) and plasma cell myeloma/multiple myeloma (PCM/MM). For both diseases the descriptions of precursor states such as monoclonal B-cell lymphocytosis and monoclonal gammopathy of uncertain significance (MGUS) have been updated including a better risk stratification model. New insights on mutational landscapes and branching evolutionary pattern were embedded as diagnostic and prognostic factors, accompanied by a revised structure for the chapter of plasma cell neoplasms. Thus, the WHO-HAEM5 leads to practical improvements of biological and clinical relevance for pathologists, clinicians, geneticists and scientists in the field of lymphoid malignancies. The present review gives an overview on the landscape of genetic alterations in CLL and plasma cell neoplasms with a focus on their impact on classification and treatment.
Collapse
Affiliation(s)
- Eugen Tausch
- Ulm University Division of CLL, Department of Internal Medicine 3 Ulm Germany
| | - Cristina López
- Institut d'Investigacions Biomèdiques August Phi i Sunyer (IDIBAPS) Barcelona Spain
| | | | - Reiner Siebert
- Ulm University and Ulm University Medical Center Institute of Human Genetics Ulm Germany
| |
Collapse
|
41
|
Eichhorst B, Ten Hacken E. Special issue on chronic lymphocytic leukemia: Prognostication and therapeutic options introductory editorial. Semin Hematol 2024; 61:69-72. [PMID: 38599948 DOI: 10.1053/j.seminhematol.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Barbara Eichhorst
- Department for Internal Medicine and Center für Integrated Oncology Aachen, Bonn, Cologne, Duesseldorf, University of Cologne, Cologne, Germany.
| | - Elisa Ten Hacken
- Department of Medicine, Division of Hematology/Oncology, Weill Cornell Medicine, New York, NY.
| |
Collapse
|
42
|
Playa-Albinyana H, Arenas F, Royo R, Giró A, López-Oreja I, Aymerich M, López-Guerra M, Frigola G, Beà S, Delgado J, Garcia-Roves PM, Campo E, Nadeu F, Colomer D. Chronic lymphocytic leukemia patient-derived xenografts recapitulate clonal evolution to Richter transformation. Leukemia 2024; 38:557-569. [PMID: 38017105 PMCID: PMC10912031 DOI: 10.1038/s41375-023-02095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
Chronic lymphocytic leukemia (CLL) is a B-cell neoplasm with a heterogeneous clinical behavior. In 5-10% of patients the disease transforms into a diffuse large-B cell lymphoma known as Richter transformation (RT), which is associated with dismal prognosis. Here, we aimed to establish patient-derived xenograft (PDX) models to study the molecular features and evolution of CLL and RT. We generated two PDXs by injecting CLL (PDX12) and RT (PDX19) cells into immunocompromised NSG mice. Both PDXs were morphologically and phenotypically similar to RT. Whole-genome sequencing analysis at different time points of the PDX evolution revealed a genomic landscape similar to RT tumors from both patients and uncovered an unprecedented RT subclonal heterogeneity and clonal evolution during PDX generation. In PDX12, the transformed cells expanded from a very small subclone already present at the CLL stage. Transcriptomic analysis of PDXs showed a high oxidative phosphorylation (OXPHOS) and low B-cell receptor (BCR) signaling similar to the RT in the patients. IACS-010759, an OXPHOS inhibitor, reduced proliferation, and circumvented resistance to venetoclax. In summary, we have generated new RT-PDX models, one of them from CLL cells that mimicked the evolution of CLL to RT uncovering intrinsic features of RT cells of therapeutical value.
Collapse
MESH Headings
- Humans
- Animals
- Mice
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Heterografts
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Clonal Evolution/genetics
- Prognosis
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
Collapse
Affiliation(s)
- Heribert Playa-Albinyana
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Fabian Arenas
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
| | - Romina Royo
- Barcelona Supercomputing Center (BSC), Barcelona, Spain
| | - Ariadna Giró
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Irene López-Oreja
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain
| | - Marta Aymerich
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain
| | - Mònica López-Guerra
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain
| | - Gerard Frigola
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain
- Molecular Pathology of Lymphoid Neoplasms Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sílvia Beà
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain
- Molecular Pathology of Lymphoid Neoplasms Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Julio Delgado
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Hematology Department, Hospital Clínic, Barcelona, Spain
- Lymphoid Neoplasms Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo M Garcia-Roves
- University of Barcelona, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Barcelona, Spain
| | - Elías Campo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain
- Molecular Pathology of Lymphoid Neoplasms Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ferran Nadeu
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain
- Molecular Pathology of Lymphoid Neoplasms Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Dolors Colomer
- Experimental Therapeutics in Lymphoid Malignancies Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Barcelona, Spain.
- University of Barcelona, Barcelona, Spain.
- Hematopathology Section, Pathology Department, Hospital Clínic, Barcelona, Spain.
| |
Collapse
|
43
|
Ong SY, Wang L. Leveraging genomics, transcriptomics and epigenomics to understand chemoimmunotherapy resistance in chronic lymphocytic leukemia. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:7. [PMID: 38434768 PMCID: PMC10905154 DOI: 10.20517/cdr.2023.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Patients with chronic lymphocytic leukemia (CLL) have differing clinical outcomes. Recent advances integrating multi-omic data have uncovered molecular subtypes in CLL with different prognostic implications and may allow better prediction of therapy response. While finite-duration chemoimmunotherapy (CIT) has enabled deep responses and prolonged duration of responses in the past, the advent of novel targeted therapy for the treatment of CLL has dramatically changed the therapeutic landscape. In this review, we discuss the latest genomic, transcriptomic, and epigenetic alterations regarded as major drivers of resistance to CIT in CLL. Further advances in genomic medicine will allow for better prediction of response to therapy and provide the basis for rational selection of therapy for long-term remissions with minimal toxicity.
Collapse
Affiliation(s)
- Shin Yeu Ong
- Department of Hematology/Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Hematology, Singapore General Hospital, Singapore 169608, Singapore
| | - Lili Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
44
|
Abstract
Lymphoid neoplasms represent a heterogeneous group of disease entities and subtypes with markedly different molecular and clinical features. Beyond genetic alterations, lymphoid tumors also show widespread epigenomic changes. These severely affect the levels and distribution of DNA methylation, histone modifications, chromatin accessibility, and three-dimensional genome interactions. DNA methylation stands out as a tracer of cell identity and memory, as B cell neoplasms show epigenetic imprints of their cellular origin and proliferative history, which can be quantified by an epigenetic mitotic clock. Chromatin-associated marks are informative to uncover altered regulatory regions and transcription factor networks contributing to the development of distinct lymphoid tumors. Tumor-intrinsic epigenetic and genetic aberrations cooperate and interact with microenvironmental cells to shape the transcriptome at different phases of lymphoma evolution, and intraclonal heterogeneity can now be characterized by single-cell profiling. Finally, epigenetics offers multiple clinical applications, including powerful diagnostic and prognostic biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Martí Duran-Ferrer
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain;
| | - José Ignacio Martín-Subero
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain;
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Departamento de Fundamentos Clínicos, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
45
|
Tsagiopoulou M, Gut IG. Machine learning and multi-omics data in chronic lymphocytic leukemia: the future of precision medicine? Front Genet 2024; 14:1304661. [PMID: 38283149 PMCID: PMC10811210 DOI: 10.3389/fgene.2023.1304661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024] Open
Abstract
Chronic lymphocytic leukemia is a complex and heterogeneous hematological malignancy. The advance of high-throughput multi-omics technologies has significantly influenced chronic lymphocytic leukemia research and paved the way for precision medicine approaches. In this review, we explore the role of machine learning in the analysis of multi-omics data in this hematological malignancy. We discuss recent literature on different machine learning models applied to single omic studies in chronic lymphocytic leukemia, with a special focus on the potential contributions to precision medicine. Finally, we highlight the recently published machine learning applications in multi-omics data in this area of research as well as their potential and limitations.
Collapse
Affiliation(s)
| | - Ivo G. Gut
- Centro Nacional de Analisis Genomico (CNAG), Barcelona, Spain
- Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
46
|
Sánchez Suárez MDM, Martín Roldán A, Alarcón-Payer C, Rodríguez-Gil MÁ, Poquet-Jornet JE, Puerta Puerta JM, Jiménez Morales A. Treatment of Chronic Lymphocytic Leukemia in the Personalized Medicine Era. Pharmaceutics 2023; 16:55. [PMID: 38258066 PMCID: PMC10818903 DOI: 10.3390/pharmaceutics16010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/26/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
Chronic lymphocytic leukemia is a lymphoproliferative disorder marked by the expansion of monoclonal, mature CD5+CD23+ B cells in peripheral blood, secondary lymphoid tissues, and bone marrow. The disease exhibits significant heterogeneity, with numerous somatic genetic alterations identified in the neoplastic clone, notably mutated TP53 and immunoglobulin heavy chain mutational statuses. Recent studies emphasize the pivotal roles of genetics and patient fragility in treatment decisions. This complexity underscores the need for a personalized approach, tailoring interventions to individual genetic profiles for heightened efficacy. The era of personalized treatment in CLL signifies a transformative shift, holding the potential for improved outcomes in the conquest of this intricate hematologic disorder. This review plays a role in elucidating the evolving CLL treatment landscape, encompassing all reported genetic factors. Through a comprehensive historical analysis, it provides insights into the evolution of CLL management. Beyond its retrospective nature, this review could be a valuable resource for clinicians, researchers, and stakeholders, offering a window into the latest advancements. In essence, it serves as a dynamic exploration of our current position and the promising prospects on the horizon.
Collapse
Affiliation(s)
- María Del Mar Sánchez Suárez
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| | - Alicia Martín Roldán
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| | - Carolina Alarcón-Payer
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| | - Miguel Ángel Rodríguez-Gil
- Unidad de Gestión Clínica Hematología y Hemoterapia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.Á.R.-G.); (J.M.P.P.)
| | | | - José Manuel Puerta Puerta
- Unidad de Gestión Clínica Hematología y Hemoterapia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.Á.R.-G.); (J.M.P.P.)
| | - Alberto Jiménez Morales
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, 18014 Granada, Granada, Spain; (M.D.M.S.S.); (A.M.R.); (A.J.M.)
| |
Collapse
|
47
|
Ten Hacken E, Gruber M, Hernández-Sánchez M, Hoffmann GB, Baranowski K, Redd RA, Clement K, Livak K, Wu CJ. Generation of mouse models carrying B cell restricted single or multiplexed loss-of-function mutations through CRISPR-Cas9 gene editing. STAR Protoc 2023; 4:102165. [PMID: 37729058 PMCID: PMC10510057 DOI: 10.1016/j.xpro.2023.102165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/15/2023] [Accepted: 02/16/2023] [Indexed: 09/22/2023] Open
Abstract
Here, we present a protocol to generate B cell restricted mouse models of loss-of-function genetic drivers typical of lymphoproliferative disorders, using stem cell engineering of murine strains carrying B cell restricted Cas9 expression. We describe steps for preparing lentivirus expressing sgRNA-mCherry, isolating hematopoietic stem and progenitor cells, and in vitro transduction. We then detail the transplantation of engineered cells into recipient mice and verification of gene edits. These mouse models represent versatile platforms to model complex disease traits typical of lymphoproliferative disorders. For complete details on the use and execution of this protocol, please refer to ten Hacken et al.,1 ten Hacken et al.,2 and ten Hacken et al.3.
Collapse
Affiliation(s)
- Elisa Ten Hacken
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Michaela Gruber
- CEMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - María Hernández-Sánchez
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Kaitlyn Baranowski
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Robert A Redd
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
| | - Kendell Clement
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Molecular Pathology Unit, Center for Cancer Research and Center for Computational and Integrative Biology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kenneth Livak
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA; Translational Immunogenomics Laboratory, Dana Farber Cancer Institute, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
48
|
Syrykh C, Pons-Brun B, Russiñol N, Playa-Albinyana H, Baumann T, Duran-Ferrer M, Kulis M, Carbó-Meix A, Mozas P, Alcoceba M, González M, Navarro-Bailón A, Colado E, Payer ÁR, Aymerich M, Terol MJ, Lu J, Knisbacher BA, Hahn CK, Ruiz-Gaspà S, Enjuanes A, Wu CJ, Getz G, Zenz T, López-Guillermo A, Martín-Subero JI, Colomer D, Delgado J, Campo E, Nadeu F. IGLV3-21R110 mutation has prognostic value in patients with treatment-naive chronic lymphocytic leukemia. Blood Adv 2023; 7:7384-7391. [PMID: 37505099 PMCID: PMC10726246 DOI: 10.1182/bloodadvances.2023010132] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/29/2023] Open
Affiliation(s)
- Charlotte Syrykh
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Pathology, Institut Universitaire du Cancer, CHU de Toulouse, Toulouse, France
| | - Berta Pons-Brun
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Núria Russiñol
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Heribert Playa-Albinyana
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | | | - Martí Duran-Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Marta Kulis
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Anna Carbó-Meix
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Pablo Mozas
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
| | - Miguel Alcoceba
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Biología Molecular e Histocompatibilidad, Instituto de Investigación Biomédica de Salamanca-Hospital Universitario, Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer, University of Salamanca-Spanish National Research Council, Salamanca, Spain
| | - Marcos González
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Biología Molecular e Histocompatibilidad, Instituto de Investigación Biomédica de Salamanca-Hospital Universitario, Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer, University of Salamanca-Spanish National Research Council, Salamanca, Spain
| | - Almudena Navarro-Bailón
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Biología Molecular e Histocompatibilidad, Instituto de Investigación Biomédica de Salamanca-Hospital Universitario, Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer, University of Salamanca-Spanish National Research Council, Salamanca, Spain
| | - Enrique Colado
- Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Ángel R. Payer
- Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Marta Aymerich
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
| | - María J. Terol
- Servicio de Hematología, Hospital Clínico Universitario, Instituto de Investigación Sanitaria (INCLIVA), Universidad de Valencia, Valencia, Spain
| | - Junyan Lu
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Binyamin A. Knisbacher
- The Broad Institute of MIT and Harvard, Cambridge, MA
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Cynthia K. Hahn
- The Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Sílvia Ruiz-Gaspà
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Anna Enjuanes
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Catherine J. Wu
- The Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Gad Getz
- The Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital and University of Zürich, Zurich, Switzerland
| | - Armando López-Guillermo
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - José I. Martín-Subero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Dolors Colomer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Julio Delgado
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Elías Campo
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Hospital Clínic of Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Ferran Nadeu
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| |
Collapse
|
49
|
Hortal AM, Villanueva A, Arellano I, Prieto C, Mendoza P, Bustelo XR, Alarcón B. Mice Overexpressing Wild-Type RRAS2 Are a Novel Model for Preclinical Testing of Anti-Chronic Lymphocytic Leukemia Therapies. Cancers (Basel) 2023; 15:5817. [PMID: 38136362 PMCID: PMC10742337 DOI: 10.3390/cancers15245817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
B-cell chronic lymphocytic leukemia (B-CLL) is the most common type of leukemia in the Western world. Mutation in different genes, such as TP53 and ATM, and deletions at specific chromosomic regions, among which are 11q or 17p, have been described to be associated to worse disease prognosis. Recent research from our group has demonstrated that, contrary to what is the usual cancer development process through missense mutations, B-CLL is driven by the overexpression of the small GTPase RRAS2 in its wild-type form without activating mutations. Some mouse models of this disease have been developed to date and are commonly used in B-CLL research, but they present different disadvantages such as the long waiting period until the leukemia fully develops, the need to do cell engraftment or, in some cases, the fact that the model does not recapitulate the alterations found in human patients. We have recently described Rosa26-RRAS2fl/flxmb1-Cre as a new mouse model of B-CLL with a full penetrance of the disease. In this work, we have validated this mouse model as a novel tool for the development of new therapies for B-CLL, by testing two of the most broadly applied targeted agents: ibrutinib and venetoclax. This also opens the door to new targeted agents against R-RAS2 itself, an approach not yet explored in the clinic.
Collapse
Affiliation(s)
- Alejandro M. Hortal
- Immune System Development and Function Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.V.); (I.A.); (C.P.); (P.M.)
| | - Ana Villanueva
- Immune System Development and Function Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.V.); (I.A.); (C.P.); (P.M.)
| | - Irene Arellano
- Immune System Development and Function Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.V.); (I.A.); (C.P.); (P.M.)
| | - Cristina Prieto
- Immune System Development and Function Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.V.); (I.A.); (C.P.); (P.M.)
| | - Pilar Mendoza
- Immune System Development and Function Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.V.); (I.A.); (C.P.); (P.M.)
| | - Xosé R. Bustelo
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer and Centro de Investigación Biomédica en Red de Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, 37007 Salamanca, Spain;
| | - Balbino Alarcón
- Immune System Development and Function Program, Centro Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; (A.V.); (I.A.); (C.P.); (P.M.)
| |
Collapse
|
50
|
Grau M, López C, Martín-Subero JI, Beà S. Cytogenomics of B-cell non-Hodgkin lymphomas: The "old" meets the "new". Best Pract Res Clin Haematol 2023; 36:101513. [PMID: 38092483 DOI: 10.1016/j.beha.2023.101513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 12/18/2023]
Abstract
For the routine diagnosis of haematological neoplasms an integrative approach is used considering the morphology, and the immunophenotypic, and molecular features of the tumor sample, along with clinical information. The identification and characterization of recurrent chromosomal aberrations mainly detected by conventional and molecular cytogenetics in the tumor cells has a major impact on the classification of lymphoid neoplasms. Some of the B-cell non-Hodgkin lymphomas are characterized by particular chromosomal aberrations, highlighting the relevance of conventional and molecular cytogenetic studies in their diagnosis and prognosis. In the current genomics era, next generation sequencing provides relevant information as the mutational profiles of haematological malignancies, improving their classification and also the clinical management of the patients. In addition, other new technologies have emerged recently, such as the optical genome mapping, which can overcome some of the limitations of conventional and molecular cytogenetics and may become more widely used in the cytogenetic laboratories in the upcoming years. Moreover, epigenetic alterations may complement genetic changes for a deeper understanding of the pathogenesis underlying B-cell neoplasms and a more precise risk-based patient stratification. Overall, here we describe the current state of the genomic data integrating chromosomal rearrangements, copy number alterations, and somatic variants, as well as a succinct overview of epigenomic changes, which altogether constitute a comprehensive diagnostic approach in B-cell non-Hodgkin lymphomas.
Collapse
Affiliation(s)
- Marta Grau
- Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Cristina López
- Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Spain
| | - José Ignacio Martín-Subero
- Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Sílvia Beà
- Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Universitat de Barcelona, Spain; Hematopathology Section, Pathology Department, Hospital Clínic Barcelona, Barcelona, Spain.
| |
Collapse
|