101
|
Huo YJ, Xu PP, Wang L, Zhong HJ, Fu D, Shi Q, Cheng S, Wang S, Zhang MC, Zhao WL. Dynamic change of soluble interleukin-2 receptor distinguished molecular heterogeneity and microenvironment alterations in diffuse large B-cell lymphoma. Biomark Res 2022; 10:51. [PMID: 35879731 PMCID: PMC9316360 DOI: 10.1186/s40364-022-00401-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/20/2022] [Indexed: 12/05/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive lymphoma with variable clinical outcomes and prediction of prognosis remains important for long-term remission. We performed serial serum soluble interleukin-2 receptor (sIL-2R) measurement pretreatment and before each cycle of the treatment in 599 patients with de novo DLBCL. Genomic and transcriptomic features were analyzed by 223 DNA- and 227 RNA-sequencing, respectively. Applying the cut-off value to sIL-2R pretreatment and cycle 2 (C2) level, patients were classified into FINE subtype (pretreatment low level) with good prognosis, RES subtype (pretreatment high level and C2 low level) with intermediate prognosis, and RET subtype (pretreatment high level and C2 high level) with poor prognosis, independent of International Prognostic Index. In “others” genetic subtype, dynamic change of sIL-2R showed prognostic significance and genetic features. Compared with FINE subtype, RES subtype had increased ARID1A and MYD88 mutations, and RET subtype had increased KMT2D, LYN and SOCS1 mutations. RES and RET subtypes showed significant enrichment in oncogenic pathways, such as ERK, NF-κB, JAK-STAT, and immune-associated pathways. As for tumor microenvironment, RES subtype exhibited increased recruiting activity of CD8 + T, T helper 1, and natural killer cells, and RET subtype with increased recruiting activity of CD4 + T and regulatory T cells in silico. There was a positive correlation between transcripts of IL-2R and immune checkpoint expressions including PD-1 and CTLA-4. Our findings identified that dynamic change of sIL-2R, with this simple and easy detection method in peripheral blood, had long-term prognostic effect and specific relation to microenvironment alterations in DLBCL.
Collapse
Affiliation(s)
- Yu-Jia Huo
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China.,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Hui-Juan Zhong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Di Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Qing Shi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Shu Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Shuo Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Mu-Chen Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Shanghai, 200025, China. .,Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
102
|
Loeffler-Wirth H, Kreuz M, Schmidt M, Ott G, Siebert R, Binder H. Classifying Germinal Center Derived Lymphomas-Navigate a Complex Transcriptional Landscape. Cancers (Basel) 2022; 14:3434. [PMID: 35884496 PMCID: PMC9321060 DOI: 10.3390/cancers14143434] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Classification of lymphoid neoplasms is based mainly on histologic, immunologic, and (rarer) genetic features. It has been supplemented by gene expression profiling (GEP) in the last decade. Despite the considerable success, particularly in associating lymphoma subtypes with specific transcriptional programs and classifier signatures of up- or downregulated genes, competing molecular classifiers were often proposed in the literature by different groups for the same classification tasks to distinguish, e.g., BL versus DLBCL or different DLBCL subtypes. Moreover, rarer sub-entities such as MYC and BCL2 "double hit lymphomas" (DHL), IRF4-rearranged large cell lymphoma (IRF4-LCL), and Burkitt-like lymphomas with 11q aberration pattern (mnBLL-11q) attracted interest while their relatedness regarding the major classes is still unclear in many respects. We explored the transcriptional landscape of 873 lymphomas referring to a wide spectrum of subtypes by applying self-organizing maps (SOM) machine learning. The landscape reveals a continuum of transcriptional states activated in the different subtypes without clear-cut borderlines between them and preventing their unambiguous classification. These states show striking parallels with single cell gene expression of the active germinal center (GC), which is characterized by the cyclic progression of B-cells. The expression patterns along the GC trajectory are discriminative for distinguishing different lymphoma subtypes. We show that the rare subtypes take intermediate positions between BL, DLBCL, and FL as considered by the 5th edition of the WHO classification of haemato-lymphoid tumors in 2022. Classifier gene signatures extracted from these states as modules of coregulated genes are competitive with literature classifiers. They provide functional-defined classifiers with the option of consenting redundant classifiers from the literature. We discuss alternative classification schemes of different granularity and functional impact as possible avenues toward personalization and improved diagnostics of GC-derived lymphomas.
Collapse
Affiliation(s)
- Henry Loeffler-Wirth
- Interdisciplinary Centre for Bioinformatics, University Leipzig (IZBI), 04107 Leipzig, Germany; (H.L.-W.); (M.S.)
| | - Markus Kreuz
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany;
| | - Maria Schmidt
- Interdisciplinary Centre for Bioinformatics, University Leipzig (IZBI), 04107 Leipzig, Germany; (H.L.-W.); (M.S.)
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany;
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, 89073 Ulm, Germany;
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, University Leipzig (IZBI), 04107 Leipzig, Germany; (H.L.-W.); (M.S.)
| |
Collapse
|
103
|
Simonson PD, Valencia I, Patel SS. Tyramide-conjugated DNA barcodes enable signal amplification for multiparametric CODEX imaging. Commun Biol 2022; 5:627. [PMID: 35754060 PMCID: PMC9234042 DOI: 10.1038/s42003-022-03558-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 06/06/2022] [Indexed: 01/09/2023] Open
Abstract
Multiparametric imaging allows researchers to measure the expression of many biomarkers simultaneously, allowing detailed characterization of cell microenvironments. One such technique, CODEX, allows fluorescence imaging of >30 proteins in a single tissue section. In the commercial CODEX system, primary antibodies are conjugated to DNA barcodes. This modification can result in antibody dysfunction, and development of a custom antibody panel can be very costly and time consuming as trial and error of modified antibodies proceeds. To address these challenges, we developed novel tyramide-conjugated DNA barcodes that can be used with primary antibodies via peroxidase-conjugated secondary antibodies. This approach results in signal amplification and imaging without the need to conjugate primary antibodies. When combined with commercially available barcode-conjugated primary antibodies, we can very quickly develop working antibody panels. We also present methods to perform antibody staining using a commercially available automated tissue stainer and in situ hybridization imaging on a CODEX platform. Future work will include application of the combined tyramide-based and regular CODEX approach to image specific tumors with their immune cell infiltrates, including biomarkers that are currently difficult to image by regular CODEX.
Collapse
Affiliation(s)
- Paul D Simonson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Itzel Valencia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sanjay S Patel
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
104
|
CAR T-cell Therapy in Highly-Aggressive B-Cell Lymphoma: Emerging Biological and Clinical Insights. Blood 2022; 140:1461-1469. [PMID: 35560330 DOI: 10.1182/blood.2022016226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Recently, significant progress has been made in identifying novel therapies, beyond conventional immunochemotherapy strategies, with efficacy in B-cell lymphomas. One such approach involves targeting the CD19 antigen on B-cells with autologous-derived chimeric antigen receptor (CAR) cells. This strategy is highly effective in patients with relapsed and refractory diffuse large B-cell lymphoma (DLBCL) as evidenced by recent regulatory approvals. Recent reports suggest that this is an effective strategy for high-grade B-cell. The biological underpinnings of these entities and how they overlap with each other and DLBCL continue to be areas of intense investigation. Therefore, as more experience with CAR T-cell approaches is examined, it is interesting to consider how both tumor-cell specific and microenvironment factors that define these highly aggressive subsets influence susceptibility to this approach.
Collapse
|
105
|
L'Imperio V, Morello G, Vegliante MC, Cancila V, Bertolazzi G, Mazzara S, Belmonte B, Mangogna A, Balzarini P, Corral L, Lopez G, Di Napoli A, Facchetti F, Pagni F, Tripodo C. Spatial transcriptome of a germinal center plasmablastic burst hints at MYD88/CD79B mutants-enriched diffuse large B-cell lymphomas. Eur J Immunol 2022; 52:1350-1361. [PMID: 35554927 PMCID: PMC9546146 DOI: 10.1002/eji.202149746] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022]
Abstract
The GC reaction results in the selection of B cells acquiring effector Ig secreting ability by progressing toward plasmablastic differentiation. This transition is associated with exclusion from the GC microenvironment. The aberrant expansion of plasmablastic elements within the GC fringes configures an atypical condition, the biological characteristics of which have not been defined yet. We investigated the in situ immunophenotypical and transcriptional characteristics of a nonclonal germinotropic expansion of plasmablastic elements (GEx) occurring in the tonsil of a young patient. Compared to neighboring GC and perifollicular regions, the GEx showed a distinctive signature featuring key regulators of plasmacytic differentiation, cytokine signaling, and cell metabolism. The GEx signature was tested in the setting of diffuse large B‐cell lymphoma (DLBCL) as a prototypical model of lymphomagenesis encompassing transformation at different stages of GC and post‐GC functional differentiation. The signature outlined DLBCL clusters with different immune microenvironment composition and enrichment in genetic subtypes. This report represents the first insight into the transcriptional features of a germinotropic plasmablastic burst, shedding light into the molecular hallmarks of B cells undergoing plasmablastic differentiation and aberrant expansion within the noncanonical setting of the GC microenvironment.
Collapse
Affiliation(s)
- Vincenzo L'Imperio
- Department of Medicine and Surgery, University of Milano-Bicocca, Pathology, San Gerardo Hospital, Via G.B. Pergolesi 33, Monza, Italy
| | - Gaia Morello
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | | | - Valeria Cancila
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Giorgio Bertolazzi
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Saveria Mazzara
- Division of Diagnostic Haematopathology, European Institute of Oncology, Milan, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) "Burlo Garofolo", Trieste, 34137, Italy
| | - Piera Balzarini
- Department of Molecular and Translational Medicine, University of Brescia, Piazzale Spedali Civili 1, Brescia, 25123, Italy
| | - Lilia Corral
- Centro Ricerca Tettamanti, Pediatric Clinic, University of Milan Bicocca, San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Gianluca Lopez
- Pathology Unit, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Arianna Di Napoli
- Pathology Unit, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | | | - Fabio Pagni
- Department of Medicine and Surgery, University of Milano-Bicocca, Pathology, San Gerardo Hospital, Via G.B. Pergolesi 33, Monza, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy.,the FIRC Institute of Molecular Oncology, Tumor and Microenvironment Histopathology Unit, IFOM, Milan, Italy
| |
Collapse
|
106
|
Zhao Y, Xu H, Zhang M, Li L. Single-Cell RNA-Seq and Bulk RNA-Seq Reveal Intratumoral Heterogeneity and Tumor Microenvironment Characteristics in Diffuse Large B-Cell Lymphoma. Front Genet 2022; 13:881345. [PMID: 35601491 PMCID: PMC9116505 DOI: 10.3389/fgene.2022.881345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Diffuse large B-cell lymphoma (DLBCL) is the most common histologic subtype of non-Hodgkin’s lymphoma (NHL) with highly heterogeneous genetic and phenotypic features. Therefore, a comprehensive understanding of cellular diversity and intratumoral heterogeneity is essential to elucidate the mechanisms driving DLBCL progression and to develop new therapeutic approaches. Methods: We analyzed single-cell transcriptomic data from 2 reactive lymph node tissue samples and 2 DLBCL lymph node biopsy tissue samples to explore the transcriptomic landscape of DLBCL. In addition, we constructed a prognostic model based on the genes obtained from differential analysis. Results: Based on gene expression profiles at the single cell level, we identified and characterized different subpopulations of malignant and immune cells. Malignant cells exhibited a high degree of inter-tumor heterogeneity. Tumor-infiltrating regulatory CD4+ T cells showed highly immunosuppressive properties and exhausted cytotoxic CD8+ T cells were highly expressed with markers of exhaustion. Cell communication analysis identified complex interactions between malignant cells and other cell subpopulations. In addition, the prognostic model we constructed allows for monitoring the prognosis of DLBCL patients. Conclusion: This study provides an in-depth dissection of the transcriptional features of malignant B cells and tumor microenvironment (TME) in DLBCL and provides new insights into the tumor heterogeneity of DLBCL.
Collapse
|
107
|
Molecular Diagnostic Review of Diffuse Large B-Cell Lymphoma and Its Tumor Microenvironment. Diagnostics (Basel) 2022; 12:diagnostics12051087. [PMID: 35626243 PMCID: PMC9139291 DOI: 10.3390/diagnostics12051087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma. It is a clinically and morphologically heterogeneous entity that has continued to resist complete subtyping. Molecular subtyping efforts emerged in earnest with the advent of gene expression profiling (GEP). This molecular subtyping approach has continued to evolve simultaneously with others including immunohistochemistry and more modern genomic approaches. Recently, the veritable explosion of genomic data availability and evolving computational methodologies have provided additional avenues, by which further understanding and subclassification of DBLCLs is possible. The goal of this review is to provide a historical overview of the major classification timepoints in the molecular subtyping of DLBCL, from gene expression profiling to present day understanding.
Collapse
|
108
|
de Groot FA, de Groen RAL, van den Berg A, Jansen PM, Lam KH, Mutsaers PGNJ, van Noesel CJM, Chamuleau MED, Stevens WBC, Plaça JR, Mous R, Kersten MJ, van der Poel MMW, Tousseyn T, Woei-a-Jin FJSH, Diepstra A, Nijland M, Vermaat JSP. Biological and Clinical Implications of Gene-Expression Profiling in Diffuse Large B-Cell Lymphoma: A Proposal for a Targeted BLYM-777 Consortium Panel as Part of a Multilayered Analytical Approach. Cancers (Basel) 2022; 14:cancers14081857. [PMID: 35454765 PMCID: PMC9028345 DOI: 10.3390/cancers14081857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023] Open
Abstract
Gene-expression profiling (GEP) is used to study the molecular biology of lymphomas. Here, advancing insights from GEP studies in diffuse large B-cell lymphoma (DLBCL) lymphomagenesis are discussed. GEP studies elucidated subtypes based on cell-of-origin principles and profoundly changed the biological understanding of DLBCL with clinical relevance. Studies integrating GEP and next-generation DNA sequencing defined different molecular subtypes of DLBCL entities originating at specific anatomical localizations. With the emergence of high-throughput technologies, the tumor microenvironment (TME) has been recognized as a critical component in DLBCL pathogenesis. TME studies have characterized so-called "lymphoma microenvironments" and "ecotypes". Despite gained insights, unexplained chemo-refractoriness in DLBCL remains. To further elucidate the complex biology of DLBCL, we propose a novel targeted GEP consortium panel, called BLYM-777. This knowledge-based biology-driven panel includes probes for 777 genes, covering many aspects regarding B-cell lymphomagenesis (f.e., MYC signature, TME, immune surveillance and resistance to CAR T-cell therapy). Regarding lymphomagenesis, upcoming DLBCL studies need to incorporate genomic and transcriptomic approaches with proteomic methods and correlate these multi-omics data with patient characteristics of well-defined and homogeneous cohorts. This multilayered methodology potentially enhances diagnostic classification of DLBCL subtypes, prognostication, and the development of novel targeted therapeutic strategies.
Collapse
Affiliation(s)
- Fleur A. de Groot
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.d.G.); (R.A.L.d.G.)
| | - Ruben A. L. de Groen
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.d.G.); (R.A.L.d.G.)
| | - Anke van den Berg
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (J.R.P.); (A.D.)
| | - Patty M. Jansen
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - King H. Lam
- Department of Pathology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Pim G. N. J. Mutsaers
- Department of Hematology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Carel J. M. van Noesel
- Department of Pathology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands;
| | - Martine E. D. Chamuleau
- Cancer Center Amsterdam and LYMMCARE, Department of Hematology, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands; (M.E.D.C.); (M.J.K.)
| | - Wendy B. C. Stevens
- Department of Hematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Jessica R. Plaça
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (J.R.P.); (A.D.)
| | - Rogier Mous
- Department of Hematology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Marie José Kersten
- Cancer Center Amsterdam and LYMMCARE, Department of Hematology, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands; (M.E.D.C.); (M.J.K.)
| | - Marjolein M. W. van der Poel
- Department of Internal Medicine, Division of Hematology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands;
| | - Thomas Tousseyn
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | | | - Arjan Diepstra
- Department of Pathology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (J.R.P.); (A.D.)
| | - Marcel Nijland
- Department of Hematology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Joost S. P. Vermaat
- Department of Hematology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (F.A.d.G.); (R.A.L.d.G.)
- Correspondence:
| |
Collapse
|
109
|
Timmins MA, Ringshausen I. Transforming Growth Factor-Beta Orchestrates Tumour and Bystander Cells in B-Cell Non-Hodgkin Lymphoma. Cancers (Basel) 2022; 14:1772. [PMID: 35406544 PMCID: PMC8996985 DOI: 10.3390/cancers14071772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Transforming growth factor-beta (TGFB) is a critical regulator of normal haematopoiesis. Dysregulation of the TGFB pathway is associated with numerous haematological malignancies including myelofibrosis, acute myeloid leukaemia, and lymphoid disorders. TGFB has classically been seen as a negative regulator of proliferation in haematopoiesis whilst stimulating differentiation and apoptosis, as required to maintain homeostasis. Tumours frequently develop intrinsic resistant mechanisms to homeostatic TGFB signalling to antagonise its tumour-suppressive functions. Furthermore, elevated levels of TGFB enhance pathogenesis through modulation of the immune system and tumour microenvironment. Here, we review recent advances in the understanding of TGFB signalling in B-cell malignancies with a focus on the tumour microenvironment. Malignant B-cells harbour subtype-specific alterations in TGFB signalling elements including downregulation of surface receptors, modulation of SMAD signalling proteins, as well as genetic and epigenetic aberrations. Microenvironmental TGFB generates a protumoural niche reprogramming stromal, natural killer (NK), and T-cells. Increasingly, evidence points to complex bi-directional cross-talk between cells of the microenvironment and malignant B-cells. A greater understanding of intercellular communication and the context-specific nature of TGFB signalling may provide further insight into disease pathogenesis and future therapeutic strategies.
Collapse
Affiliation(s)
- Matthew A. Timmins
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospital, Cambridge CB2 0AH, UK
| | - Ingo Ringshausen
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospital, Cambridge CB2 0AH, UK
| |
Collapse
|
110
|
Martin P, Bartlett NL, Chavez JC, Reagan JL, Smith SM, LaCasce AS, Jones J, Drew J, Wu C, Mulvey E, Revuelta MV, Cerchietti L, Leonard JP. Phase 1 study of oral azacitidine (CC-486) plus R-CHOP in previously untreated intermediate- to high-risk DLBCL. Blood 2022; 139:1147-1159. [PMID: 34428285 PMCID: PMC9211445 DOI: 10.1182/blood.2021011679] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/30/2021] [Indexed: 11/20/2022] Open
Abstract
Resistance to standard immunochemotherapy remains an unmet challenge in diffuse large B-cell lymphoma (DLBCL), and aberrant DNA methylation may contribute to chemoresistance. Promising early-phase results were reported with rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) plus subcutaneous azacitidine, a hypomethylating agent. In this phase 1 study, we evaluated CC-486 (oral azacitidine) plus 6 cycles of R-CHOP in patients with previously untreated intermediate- to high-risk DLBCL or grade 3B/transformed follicular lymphoma. CC-486 doses of 100, 150, 200, or 300 mg given 7 days before cycle 1 and on days 8-21 of cycles 1-5 were evaluated; additional patients were enrolled in the expansion phase to examine preliminary efficacy. The primary objectives were to determine the safety and the maximum tolerated dose (MTD) of CC-486 in combination with R-CHOP. The most common grade 3/4 toxicities were hematologic, including neutropenia (62.7%) and febrile neutropenia (25.4%); grade 3/4 nonhematologic toxicities were uncommon (<7%). The MTD was not established; 2 patients had dose-limiting toxicities (1 with grade 4 febrile neutropenia; 1 with grade 4 prolonged neutropenia). The recommended phase 2 dose was established as 300 mg. The overall response rate was 94.9%, with 52 patients (88.1%) achieving complete responses. With a median follow-up of 28.9 months, estimated 1- and 2-year progression-free survival rates were 84.1% and 78.6%, respectively. Overall, epigenetic priming with CC-486 before R-CHOP can be delivered with acceptable safety to patients with previously untreated intermediate- to high-risk DLBCL or grade 3B/transformed follicular lymphoma. ClinicalTrials.gov: NCT02343536.
Collapse
Affiliation(s)
| | | | | | - John L Reagan
- The Warren Alpert Medical School of Brown University, Providence, RI
| | - Sonali M Smith
- University of Chicago Comprehensive Cancer Center, Chicago, IL
| | | | | | | | | | | | | | | | | |
Collapse
|
111
|
Autio M, Leivonen SK, Brück O, Karjalainen-Lindsberg ML, Pellinen T, Leppä S. Clinical Impact of Immune Cells and Their Spatial Interactions in Diffuse Large B-Cell Lymphoma Microenvironment. Clin Cancer Res 2022; 28:781-792. [PMID: 34907083 PMCID: PMC9377736 DOI: 10.1158/1078-0432.ccr-21-3140] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/24/2021] [Accepted: 12/10/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Tumor-infiltrating immune cells have prognostic significance and are attractive therapeutic targets. Yet, the clinical significance of their spatial organization and phenotype in diffuse large B-cell lymphoma (DLBCL) is unclear. EXPERIMENTAL DESIGN We characterized T cells, macrophages, and their spatial interactions by multiplex IHC (mIHC) in 178 patients with DLBCL and correlated the data with patient demographics and survival. We validated the findings on gene expression data from two external DLBCL cohorts comprising 633 patients. RESULTS Macrophage and T-cell contents divided the samples into T cell-inflamed (60%) and noninflamed (40%) subgroups. The T cell-inflamed lymphoma microenvironment (LME) was also rich in other immune cells, defining immune hot phenotype, which did not as such correlate with outcome. However, when we divided the patients according to T-cell and macrophage contents, LME characterized by high T-cell/low macrophage content or a corresponding gene signature was associated with superior survival [5-year overall survival (OS): 92.3% vs. 74.4%, P = 0.036; 5-year progression-free survival (PFS): 92.6% vs. 69.8%, P = 0.012]. High proportion of PD-L1- and TIM3-expressing CD163- macrophages in the T cell-inflamed LME defined a group of patients with poor outcome [OS: HR = 3.22, 95% confidence interval (CI), 1.63-6.37, Padj = 0.011; PFS: HR = 2.76, 95% CI, 1.44-5.28, Padj = 0.016]. Furthermore, PD-L1 and PD-1 were enriched on macrophages interacting with T cells. CONCLUSIONS Our data demonstrate that the interplay between macrophages and T cells in the DLBCL LME is immune checkpoint dependent and clinically meaningful.
Collapse
Affiliation(s)
- Matias Autio
- Research Program 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
| | - Suvi-Katri Leivonen
- Research Program 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
| | - Oscar Brück
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Sirpa Leppä
- Research Program 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.,Corresponding Author: Sirpa Leppä, University of Helsinki, Haartmaninkatu 8, Helsinki FI-00014, Finland. Phone: 358-50-427-0820; E-mail:
| |
Collapse
|
112
|
Mondello P, Ansell SM, Nowakowski GS. Immune Epigenetic Crosstalk Between Malignant B Cells and the Tumor Microenvironment in B Cell Lymphoma. Front Genet 2022; 13:826594. [PMID: 35237302 PMCID: PMC8883034 DOI: 10.3389/fgene.2022.826594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/22/2022] Open
Abstract
Epigenetic reprogramming is a hallmark of lymphomagenesis, however its role in reshaping the tumor microenvironment is still not well understood. Here we review the most common chromatin modifier mutations in B cell lymphoma and their effect on B cells as well as on T cell landscape. We will also discuss precision therapy strategies to reverse their aberrant signaling by targeting mutated proteins or counterbalance epigenetic mechanisms.
Collapse
|
113
|
Abstract
Two recent reports in the New England Journal of Medicine highlight the successful application of novel targeted therapies to improve the clinical outcomes of diffuse large B-cell lymphoma (DLBCL). These findings encourage us to pursue further mechanism-based therapeutic uses to make DLBCL curable in the era of precision medicine.
Collapse
Affiliation(s)
- Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu-Jia Huo
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
114
|
Bröske AME, Korfi K, Belousov A, Wilson S, Ooi CH, Bolen CR, Canamero M, Alcaide EG, James I, Piccione EC, Carlile DJ, Dimier N, Umaña P, Bacac M, Weisser M, Dickinson M. Pharmacodynamics and molecular correlates of response to glofitamab in relapsed/refractory non-Hodgkin lymphoma. Blood Adv 2022; 6:1025-1037. [PMID: 34941996 PMCID: PMC8945294 DOI: 10.1182/bloodadvances.2021005954] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/28/2021] [Indexed: 11/20/2022] Open
Abstract
Glofitamab, a novel CD20xCD3, T-cell-engaging bispecific antibody, exhibited single-agent activity in Study NP30179, a first-in-human, phase 1 trial in relapsed/refractory B-cell non-Hodgkin lymphoma. Preclinical studies showed that glofitamab leads to T-cell activation, proliferation, and tumor cell killing upon binding to CD20 on malignant cells. Here, we provide evidence of glofitamab's clinical activity, including pharmacodynamic profile, mode of action, and factors associated with clinical response, by evaluating biomarkers in patient samples from the dose-escalation part of this trial. Patients enrolled in Study NP30179 received single-dose obinutuzumab pretreatment (1000 mg) 7 days before IV glofitamab (5 µg-25 mg). Glofitamab treatment lasted ≤12 cycles once every 2 or 3 weeks. Blood samples were collected at predefined time points per the clinical protocol; T-cell populations were evaluated centrally by flow cytometry, and cytokine profiles were analyzed. Immunohistochemical and genomic biomarker analyses were performed on tumor biopsy samples. Pharmacodynamic modulation was observed with glofitamab treatment, including dose-dependent induction of cytokines, and T-cell margination, proliferation, and activation in peripheral blood. Gene expression analysis of pretreatment tumor biopsy samples indicated that tumor cell intrinsic factors such as TP53 signaling are associated with resistance to glofitamab, but they may also be interlinked with a diminished effector T-cell profile in resistant tumors and thus represent a poor prognostic factor per se. This integrative biomarker data analysis provides clinical evidence regarding glofitamab's mode of action, supports optimal biological dose selection, and will further guide clinical development. This trial was registered at www.clinicaltrials.gov as #NCT03075696.
Collapse
Affiliation(s)
- Ann-Marie E. Bröske
- Roche Innovation Center Munich, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Koorosh Korfi
- Roche Innovation Center Zürich, Roche Pharma Research and Early Development, Zürich, Switzerland
| | - Anton Belousov
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Sabine Wilson
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Chia-Huey Ooi
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | | | - Marta Canamero
- Roche Innovation Center Munich, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Enrique Gomez Alcaide
- Roche Innovation Center Basel, Roche Pharma Research and Early Development, Basel, Switzerland
| | - Ian James
- A4P Consulting Ltd., Sandwich, United Kingdom
| | | | - David J. Carlile
- Roche Innovation Center Welwyn, Roche Pharma Research and Early Development, Welwyn Garden City, United Kingdom; and
| | - Natalie Dimier
- Roche Innovation Center Welwyn, Roche Pharma Research and Early Development, Welwyn Garden City, United Kingdom; and
| | - Pablo Umaña
- Roche Innovation Center Zürich, Roche Pharma Research and Early Development, Zürich, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zürich, Roche Pharma Research and Early Development, Zürich, Switzerland
| | - Martin Weisser
- Roche Innovation Center Munich, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Michael Dickinson
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
115
|
Bonfiglio F, Bruscaggin A, Guidetti F, Terzi di Bergamo L, Faderl M, Spina V, Condoluci A, Bonomini L, Forestieri G, Koch R, Piffaretti D, Pini K, Pirosa MC, Cittone MG, Arribas A, Lucioni M, Ghilardi G, Wu W, Arcaini L, Baptista MJ, Bastidas G, Bea S, Boldorini R, Broccoli A, Buehler MM, Canzonieri V, Cascione L, Ceriani L, Cogliatti S, Corradini P, Derenzini E, Devizzi L, Dietrich S, Elia AR, Facchetti F, Gaidano G, Garcia JF, Gerber B, Ghia P, Gomes da Silva M, Gritti G, Guidetti A, Hitz F, Inghirami G, Ladetto M, Lopez-Guillermo A, Lucchini E, Maiorana A, Marasca R, Matutes E, Meignin V, Merli M, Moccia A, Mollejo M, Montalban C, Novak U, Oscier DG, Passamonti F, Piazza F, Pizzolitto S, Rambaldi A, Sabattini E, Salles G, Santambrogio E, Scarfò L, Stathis A, Stüssi G, Geyer JT, Tapia G, Tarella C, Thieblemont C, Tousseyn T, Tucci A, Vanini G, Visco C, Vitolo U, Walewska R, Zaja F, Zenz T, Zinzani PL, Khiabanian H, Calcinotto A, Bertoni F, Bhagat G, Campo E, De Leval L, Dirnhofer S, Pileri SA, Piris MA, Traverse-Glehen A, Tzankov A, Paulli M, Ponzoni M, Mazzucchelli L, Cavalli F, Zucca E, Rossi D. Genetic and phenotypic attributes of splenic marginal zone lymphoma. Blood 2022; 139:732-747. [PMID: 34653238 DOI: 10.1182/blood.2021012386] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/14/2021] [Indexed: 11/20/2022] Open
Abstract
Splenic marginal zone B-cell lymphoma (SMZL) is a heterogeneous clinico-biological entity. The clinical course is variable, multiple genes are mutated with no unifying mechanism, and essential regulatory pathways and surrounding microenvironments are diverse. We sought to clarify the heterogeneity of SMZL by resolving different subgroups and their underlying genomic abnormalities, pathway signatures, and microenvironment compositions to uncover biomarkers and therapeutic vulnerabilities. We studied 303 SMZL spleen samples collected through the IELSG46 multicenter international study (NCT02945319) by using a multiplatform approach. We carried out genetic and phenotypic analyses, defined self-organized signatures, validated the findings in independent primary tumor metadata and in genetically modified mouse models, and determined correlations with outcome data. We identified 2 prominent genetic clusters in SMZL, termed NNK (58% of cases, harboring NF-κB, NOTCH, and KLF2 modules) and DMT (32% of cases, with DNA-damage response, MAPK, and TLR modules). Genetic aberrations in multiple genes as well as cytogenetic and immunogenetic features distinguished NNK- from DMT-SMZLs. These genetic clusters not only have distinct underpinning biology, as judged by differences in gene-expression signatures, but also different outcomes, with inferior survival in NNK-SMZLs. Digital cytometry and in situ profiling segregated 2 basic types of SMZL immune microenvironments termed immune-suppressive SMZL (50% of cases, associated with inflammatory cells and immune checkpoint activation) and immune-silent SMZL (50% of cases, associated with an immune-excluded phenotype) with distinct mutational and clinical connotations. In summary, we propose a nosology of SMZL that can implement its classification and also aid in the development of rationally targeted treatments.
Collapse
Affiliation(s)
- Ferdinando Bonfiglio
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Alessio Bruscaggin
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Francesca Guidetti
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | | | - Martin Faderl
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Valeria Spina
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Adalgisa Condoluci
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Luisella Bonomini
- International Extranodal Lymphoma Study Group, Bellinzona, Switzerland
| | - Gabriela Forestieri
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Ricardo Koch
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Deborah Piffaretti
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Katia Pini
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Maria Cristina Pirosa
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Micol Giulia Cittone
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Alberto Arribas
- Lymphoma Genomics, Institute of Oncology Research, Bellinzona, Switzerland
| | - Marco Lucioni
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Guido Ghilardi
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Wei Wu
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Luca Arcaini
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Maria Joao Baptista
- Lymphoid Neoplasms Group, Josep Carreras Leukaemia Research Institute, Badalona, Spain
| | - Gabriela Bastidas
- Division of Hematology, Hospital Clínic i Provincial de Barcelona, Barcelona, Spain
| | - Silvia Bea
- Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) 28029, Madrid, Spain
- Pathology Department, Hospital Clínic, Barcelona University, Barcelona, Spain
| | - Renzo Boldorini
- Division of Pathology, University of Eastern Piedmont, Novara, Italy
| | - Alessandro Broccoli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia Seràgnoli, Bologna, Italy
| | - Marco Matteo Buehler
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Vincenzo Canzonieri
- Pathology Unit, CRO Aviano National Cancer Institute, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Luciano Cascione
- Lymphoma Genomics, Institute of Oncology Research, Bellinzona, Switzerland
| | - Luca Ceriani
- Clinic of Nuclear Medicine and PET-CT Centre, Imaging Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Sergio Cogliatti
- Institute of Pathology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Paolo Corradini
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Enrico Derenzini
- Onco-hematology Division, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Liliana Devizzi
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Sascha Dietrich
- Division of Hematology, University Hospital Heidelberg, Heidelberg, Germany
| | - Angela Rita Elia
- Cancer Immunotherapy, Institute of Oncology Research, Bellinzona, Switzerland
| | - Fabio Facchetti
- Department of Molecular and Translational Medicine, Pathology Unit, Spedali Civili, Brescia, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | | | - Bernhard Gerber
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Department of Hematology and Oncology, University of Zurich, Zurich, Switzerland
| | - Paolo Ghia
- Strategic Research Program on Chronic Lymphocytic Leukemia (CLL), IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Gomes da Silva
- Division of Hematology, Instituto Português de Oncologia de Lisboa, Lisbon, Portugal
| | - Giuseppe Gritti
- Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Anna Guidetti
- Division of Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Felicitas Hitz
- Division of Hematology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Marco Ladetto
- Division of Hematology, Azienda Ospedaliera SS Antonio e Biagio, Alessandria, Italy
- Dipartimento di Medicina Traslazionale, University of Eastern Piedmont, Alessandria, Italy
| | | | - Elisa Lucchini
- Division of Hematology, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Antonino Maiorana
- Division of Pathology, Universitá degli Studi di Modena e Reggio Emilia, Modena, Italy
| | - Roberto Marasca
- Hematology Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Estella Matutes
- Haematopathology Unit, Hospital Clínic i Provincial de Barcelona, Barcelona, Spain
| | | | - Michele Merli
- Division of Hematology, University of Insubria and ASST Sette Laghi, Ospedale di Circolo of Varese, Varese, Italy
| | - Alden Moccia
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Manuela Mollejo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) 28029, Madrid, Spain
- Division of Pathology, Hospital Virgen de la Salud, Toledo, Spain
| | - Carlos Montalban
- Division of Hematology, MD Anderson Cancer Center, Madrid, Spain
| | - Urban Novak
- Department of Medical Oncology and University Cancer Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David Graham Oscier
- Division of Hematology, University Hospitals Dorset, Bournemouth, United Kingdom
| | - Francesco Passamonti
- Department of Medicine and Surgery, University of Insubria and ASST Sette Laghi, Ospedale di Circolo of Varese, Varese, Italy
| | - Francesco Piazza
- Division of Hematology, Ospedale Universitario di Padova, Padova, Italy
| | - Stefano Pizzolitto
- Division of Pathology, General Hospital S Maria della Misericordia, Udine, Italy
| | - Alessandro Rambaldi
- Division of Hematology, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Elena Sabattini
- Haematopathology Unit, Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gilles Salles
- Faculté de Médecine et de Maïeutique Lyon Sud, Université de Lyon, Lyon, France
| | | | - Lydia Scarfò
- Strategic Research Program on Chronic Lymphocytic Leukemia (CLL), IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | - Anastasios Stathis
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Georg Stüssi
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
| | - Julia T Geyer
- Division of Anatomic Pathology and Clinical Pathology, Weill Cornell Medical College, New York, NY
| | - Gustavo Tapia
- Division of Pathology, Hospital Germans Trias I Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Corrado Tarella
- Onco-hematology Division, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Catherine Thieblemont
- Assistance Publique-Hôpitaux de Paris, Hopital Saint-Louis, Hemato-Oncology Unit; Université de Paris, Paris, France
| | - Thomas Tousseyn
- Department of Haematology, University Hospitals Leuven, Leuven, Belgium
| | | | - Giorgio Vanini
- Department of Medical Oncology and University Cancer Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Carlo Visco
- Department of Medicine, Section of Hematology, University of Verona, Italy
| | - Umberto Vitolo
- Candiolo Cancer Institute (FPO-IRCCS), Candiolo, Turin, Italy
| | - Renata Walewska
- Division of Hematology, University Hospitals Dorset, Bournemouth, United Kingdom
| | - Francesco Zaja
- Division of Hematology, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Thorsten Zenz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Pier Luigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia Seràgnoli, Bologna, Italy
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, Università di Bologna, Bologna, Italy
| | - Hossein Khiabanian
- Center for Systems and Computational Biology, Rutgers University, New Brunswick, NJ
| | - Arianna Calcinotto
- Cancer Immunotherapy, Institute of Oncology Research, Bellinzona, Switzerland
| | - Francesco Bertoni
- Lymphoma Genomics, Institute of Oncology Research, Bellinzona, Switzerland
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Elias Campo
- Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS)
- Pathology Department, Hospital Clínic, Barcelona University, Barcelona, Spain
| | - Laurence De Leval
- Division of Pathology, Institut Universitaire de Pathologie, Lausanne, Switzerland
| | - Stefan Dirnhofer
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Stefano A Pileri
- Haematopathology Division, European Institute of Oncology IRCCS, Milan, Italy
| | - Miguel A Piris
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC) 28029, Madrid, Spain
- Pathology Service, Fundación Jiménez Díaz, Madrid, Spain
| | | | - Alexander Tzankov
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - Marco Paulli
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo and Università degli Studi di Pavia, Pavia, Italy
| | - Maurilio Ponzoni
- Ateneo Vita-Salute San Raffaele University and Pathology Unit San Raffaele Scientific Institute, Milan, Italy
| | - Luca Mazzucchelli
- Division of Pathology, Cantonal Institute of Pathology, Locarno, Switzerland
| | - Franco Cavalli
- Institute of Oncology Research, Bellinzona, Switzerland; and
| | - Emanuele Zucca
- International Extranodal Lymphoma Study Group, Bellinzona, Switzerland
- Clinic of Medical Oncology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
- Department of Medical Oncology, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Davide Rossi
- Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Division of Hematology, Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), Lugano, Switzerland
| |
Collapse
|
116
|
Serganova I, Chakraborty S, Yamshon S, Isshiki Y, Bucktrout R, Melnick A, Béguelin W, Zappasodi R. Epigenetic, Metabolic, and Immune Crosstalk in Germinal-Center-Derived B-Cell Lymphomas: Unveiling New Vulnerabilities for Rational Combination Therapies. Front Cell Dev Biol 2022; 9:805195. [PMID: 35071240 PMCID: PMC8777078 DOI: 10.3389/fcell.2021.805195] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 11/30/2021] [Indexed: 12/24/2022] Open
Abstract
B-cell non-Hodgkin lymphomas (B-NHLs) are highly heterogenous by genetic, phenotypic, and clinical appearance. Next-generation sequencing technologies and multi-dimensional data analyses have further refined the way these diseases can be more precisely classified by specific genomic, epigenomic, and transcriptomic characteristics. The molecular and genetic heterogeneity of B-NHLs may contribute to the poor outcome of some of these diseases, suggesting that more personalized precision-medicine approaches are needed for improved therapeutic efficacy. The germinal center (GC) B-cell like diffuse large B-cell lymphomas (GCB-DLBCLs) and follicular lymphomas (FLs) share specific epigenetic programs. These diseases often remain difficult to treat and surprisingly do not respond advanced immunotherapies, despite arising in secondary lymphoid organs at sites of antigen recognition. Epigenetic dysregulation is a hallmark of GCB-DLBCLs and FLs, with gain-of-function (GOF) mutations in the histone methyltransferase EZH2, loss-of-function (LOF) mutations in histone acetyl transferases CREBBP and EP300, and the histone methyltransferase KMT2D representing the most prevalent genetic lesions driving these diseases. These mutations have the common effect to disrupt the interactions between lymphoma cells and the immune microenvironment, via decreased antigen presentation and responsiveness to IFN-γ and CD40 signaling pathways. This indicates that immune evasion is a key step in GC B-cell lymphomagenesis. EZH2 inhibitors are now approved for the treatment of FL and selective HDAC3 inhibitors counteracting the effects of CREBBP LOF mutations are under development. These treatments can help restore the immune control of GCB lymphomas, and may represent optimal candidate agents for more effective combination with immunotherapies. Here, we review recent progress in understanding the impact of mutant chromatin modifiers on immune evasion in GCB lymphomas. We provide new insights on how the epigenetic program of these diseases may be regulated at the level of metabolism, discussing the role of metabolic intermediates as cofactors of epigenetic enzymes. In addition, lymphoma metabolic adaptation can negatively influence the immune microenvironment, further contributing to the development of immune cold tumors, poorly infiltrated by effector immune cells. Based on these findings, we discuss relevant candidate epigenetic/metabolic/immune targets for rational combination therapies to investigate as more effective precision-medicine approaches for GCB lymphomas.
Collapse
Affiliation(s)
- Inna Serganova
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sanjukta Chakraborty
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Samuel Yamshon
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Yusuke Isshiki
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Ryan Bucktrout
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Ari Melnick
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Wendy Béguelin
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Roberta Zappasodi
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY, United States.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
117
|
Xu-Monette ZY, Wei L, Fang X, Au Q, Nunns H, Nagy M, Tzankov A, Zhu F, Visco C, Bhagat G, Dybkaer K, Chiu A, Tam W, Zu Y, Hsi ED, Hagemeister FB, Sun X, Han X, Go H, Ponzoni M, Ferreri AJM, Møller MB, Parsons BM, van Krieken JH, Piris MA, Winter JN, Li Y, Xu B, Albitar M, You H, Young KH. Genetic Subtyping and Phenotypic Characterization of the Immune Microenvironment and MYC/BCL2 Double Expression Reveal Heterogeneity in Diffuse Large B-cell Lymphoma. Clin Cancer Res 2022; 28:972-983. [PMID: 34980601 DOI: 10.1158/1078-0432.ccr-21-2949] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/25/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Diffuse large B-cell lymphoma (DLBCL) is molecularly and clinically heterogeneous, and can be subtyped according to genetic alterations, cell-of-origin, or microenvironmental signatures using high-throughput genomic data at the DNA or RNA level. Although high-throughput proteomic profiling has not been available for DLBCL subtyping, MYC/BCL2 protein double expression (DE) is an established prognostic biomarker in DLBCL. The purpose of this study is to reveal the relative prognostic roles of DLBCL genetic, phenotypic, and microenvironmental biomarkers. EXPERIMENTAL DESIGN We performed targeted next-generation sequencing; IHC for MYC, BCL2, and FN1; and fluorescent multiplex IHC for microenvironmental markers in a large cohort of DLBCL. We performed correlative and prognostic analyses within and across DLBCL genetic subtypes and MYC/BCL2 double expressors. RESULTS We found that MYC/BCL2 double-high-expression (DhE) had significant adverse prognostic impact within the EZB genetic subtype and LymphGen-unclassified DLBCL cases but not within MCD and ST2 genetic subtypes. Conversely, KMT2D mutations significantly stratified DhE but not non-DhE DLBCL. T-cell infiltration showed favorable prognostic effects within BN2, MCD, and DhE but unfavorable effects within ST2 and LymphGen-unclassified cases. FN1 and PD-1-high expression had significant adverse prognostic effects within multiple DLBCL genetic/phenotypic subgroups. The prognostic effects of DhE and immune biomarkers within DLBCL genetic subtypes were independent although DhE and high Ki-67 were significantly associated with lower T-cell infiltration in LymphGen-unclassified cases. CONCLUSIONS Together, these results demonstrated independent and additive prognostic effects of phenotypic MYC/BCL2 and microenvironment biomarkers and genetic subtyping in DLBCL prognostication, important for improving DLBCL classification and identifying prognostic determinants and therapeutic targets.
Collapse
Affiliation(s)
- Zijun Y Xu-Monette
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, North Carolina.
| | - Li Wei
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, North Carolina.,Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qingyan Au
- NeoGenomics Laboratories, Aliso Viejo, California
| | - Harry Nunns
- NeoGenomics Laboratories, Aliso Viejo, California
| | - Máté Nagy
- NeoGenomics Laboratories, Aliso Viejo, California
| | - Alexandar Tzankov
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Feng Zhu
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | | | - Govind Bhagat
- Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, New York
| | | | | | - Wayne Tam
- Weill Medical College of Cornell University, New York, New York
| | - Youli Zu
- The Methodist Hospital, Houston, Texas
| | | | - Fredrick B Hagemeister
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoping Sun
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xin Han
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heounjeong Go
- Asan Medical Center, Ulsan University College of Medicine, Seoul, Republic of South Korea
| | | | | | | | | | - J Han van Krieken
- Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Miguel A Piris
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Jane N Winter
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Yong Li
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Bing Xu
- The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Maher Albitar
- Genomic Testing Cooperative, LCA, Irvine, California
| | - Hua You
- Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Ken H Young
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, North Carolina. .,Duke Cancer Institute, Durham, North Carolina
| |
Collapse
|
118
|
Dumontet E, Mancini SJC, Tarte K. Bone Marrow Lymphoid Niche Adaptation to Mature B Cell Neoplasms. Front Immunol 2021; 12:784691. [PMID: 34956214 PMCID: PMC8694563 DOI: 10.3389/fimmu.2021.784691] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/23/2021] [Indexed: 01/08/2023] Open
Abstract
B-cell non-Hodgkin lymphoma (B-NHL) evolution and treatment are complicated by a high prevalence of relapses primarily due to the ability of malignant B cells to interact with tumor-supportive lymph node (LN) and bone marrow (BM) microenvironments. In particular, progressive alterations of BM stromal cells sustain the survival, proliferation, and drug resistance of tumor B cells during diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), and chronic lymphocytic leukemia (CLL). The current review describes how the crosstalk between BM stromal cells and lymphoma tumor cells triggers the establishment of the tumor supportive niche. DLBCL, FL, and CLL display distinct patterns of BM involvement, but in each case tumor-infiltrating stromal cells, corresponding to cancer-associated fibroblasts, exhibit specific phenotypic and functional features promoting the recruitment, adhesion, and survival of tumor cells. Tumor cell-derived extracellular vesicles have been recently proposed as playing a central role in triggering initial induction of tumor-supportive niches, notably within the BM. Finally, the disruption of the BM stroma reprogramming emerges as a promising therapeutic option in B-cell lymphomas. Targeting the crosstalk between BM stromal cells and malignant B cells, either through the inhibition of stroma-derived B-cell growth factors or through the mobilization of clonal B cells outside their supportive BM niche, should in particular be further evaluated as a way to avoid relapses by abrogating resistance niches.
Collapse
Affiliation(s)
- Erwan Dumontet
- Univ Rennes, Institut National de la Santé et de la Recherche Médicale (INSERM), Établissement Français du Sang (EFS) Bretagne, Unité Mixte de Recherche (UMR) U1236, Rennes, France.,CHU Rennes, Pôle de Biologie, Rennes, France
| | - Stéphane J C Mancini
- Univ Rennes, Institut National de la Santé et de la Recherche Médicale (INSERM), Établissement Français du Sang (EFS) Bretagne, Unité Mixte de Recherche (UMR) U1236, Rennes, France
| | - Karin Tarte
- Univ Rennes, Institut National de la Santé et de la Recherche Médicale (INSERM), Établissement Français du Sang (EFS) Bretagne, Unité Mixte de Recherche (UMR) U1236, Rennes, France.,CHU Rennes, Pôle de Biologie, Rennes, France
| |
Collapse
|
119
|
Abstract
The activated B-cell (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) has an aggressive course and is associated with poor prognosis in the relapsed or refractory setting. ABC-DLBCL is characterized by chronic active signaling of NF-κB, which is dependent on the CARD11-BCL10-MALT1 (CBM) complex. MALT1 is a key effector of the CBM complex and activates canonical NF-κB and AP-1 among other transcription factors via distinct protease and scaffold functions. There is therefore growing interest in therapeutic targeting of MALT1 for B-cell malignancies. Here, we review recent advances in therapeutic targeting of MALT1 for ABC-DLBCL. Covalent and allosteric MALT1 protease inhibitors have been developed which inhibit growth of ABC-DLBCL in preclinical models, and two clinical MALT1 protease inhibitors are being developed in phase I clinical trials. Importantly, these compounds can overcome resistance to BTK inhibitors in preclinical models. Alternative compounds blocking the scaffold effect of MALT1 are also in early preclinical development. Blockade of MALT1 protease activity may have important implications for anti-lymphoma immunity by increasing immunogenicity of ABC-DLBCL cells and also by potentiating anti-lymphoma activity of other immune cells in the lymphoma microenvironment. Together, early data suggest that MALT1 is a promising target for ABC-DLBCL and possibly other B-cell malignancies, and can have lymphoma cell-intrinsic as well as immune-mediated therapeutic effects.
Collapse
Affiliation(s)
- Madhav R Seshadri
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ari M Melnick
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
120
|
Mu S, Shi D, Ai L, Fan F, Peng F, Sun C, Hu Y. International Prognostic Index-Based Immune Prognostic Model for Diffuse Large B-Cell Lymphoma. Front Immunol 2021; 12:732006. [PMID: 34745101 PMCID: PMC8569825 DOI: 10.3389/fimmu.2021.732006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/30/2021] [Indexed: 11/22/2022] Open
Abstract
Background The International Prognostic Index (IPI) is widely used to discriminate the prognosis of patients with diffuse large B-cell lymphoma (DLBCL). However, there is a significant need to identify novel valuable biomarkers in the context of targeted therapy, such as immune checkpoint blockade (ICB). Methods Gene expression data and clinical DLBCL information were obtained from The Cancer Genome Atlas and Gene Expression Omnibus datasets. A total of 371 immune-related genes in DLBCL patients associated with different IPI risk groups were identified by weighted gene co-expression network analysis, and eight genes were selected to construct an IPI-based immune prognostic model (IPI-IPM). Subsequently, we analyzed the somatic mutation and transcription profiles of the IPI-IPM subgroups as well as the potential clinical response to immune checkpoint blockade (ICB) in IPI-IPM subgroups. Results The IPI-IPM was constructed based on the expression of CMBL, TLCD3B, SYNDIG1, ESM1, EPHA3, HUNK, PTX3, and IL12A, where high-risk patients had worse overall survival than low-risk patients, consistent with the results in the independent validation cohorts. The comprehensive results showed that high IPI-IPM risk scores were correlated with immune-related signaling pathways, high KMT2D and CD79B mutation rates, and upregulation of inhibitory immune checkpoints, including PD-L1, BTLA, and SIGLEC7, indicating a greater potential response to ICB therapy. Conclusion The IPI-IPM has independent prognostic significance for DLBCL patients, which provides an immunological perspective to elucidate the mechanisms of tumor progression and sheds light on the development of immunotherapy for DLBCL.
Collapse
Affiliation(s)
- Shidai Mu
- Institution of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Deyao Shi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lisha Ai
- Institution of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengjuan Fan
- Institution of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Peng
- Institution of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Sun
- Institution of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institution of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
121
|
Deng Y, Su W, Zhu J, Ji H, Zhou X, Geng J, Zhu J, Zhang Q. Helicobacter pylori infection disturbs the tumor immune microenvironment and is associated with a discrepant prognosis in gastric de novo diffuse large B-cell lymphoma. J Immunother Cancer 2021; 9:jitc-2021-002947. [PMID: 34645670 PMCID: PMC8515460 DOI: 10.1136/jitc-2021-002947] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gastric diffuse large B-cell lymphoma (gDLBCL) related to Helicobacter pylori infection exhibits a wide spectrum of prognosis, and the tumor immune microenvironment (TIME) affects tumor progression. However, there are few studies on the correlation between prognosis and changes of TIME induced by H. pylori infection in de novo gDLBCL. METHODS A retrospective study was performed to determine the prognostic value of TIME related to H. pylori infection in de novo gDLBCL. A total of 252 patients were included and have been treated with standard rituximab to cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy or other similar regimens in addition to H. pylori eradication (HPE). All patients were stratified by H. pylori infection, HPE efficacy, and preliminary TIME evaluation using conventional criteria. Statistical analyses were conducted. To assess the mechanism, 30 subjects were assessed for H. pylori infection. The components and spatial distributions of TIME were analyzed. RESULTS The median follow-up of the 252 patients was 66.6 months (range 0.7-119.2), and the 5-year overall survival (OS) was 78.0%. A total of 109 H. pylori-positive cases with pathological complete remission and high tumor-infiltrating T lymphocytes (cohort 1) had significantly higher 5-year progression-free survival (88.1% vs 70.5%, p<0.001) and OS (89.2% vs 76.6%, p<0.001) than the other 143 patients (cohort 2). Among 30 patients, 19 were cytotoxin-associated gene A-marked as the cohort 1 subset. Compared with cohort 2, cohort 1 exhibited increased inflammatory factors (tumor necrosis factor-α, interferon γ, etc) and decreased immunosuppressive components (PD-L1, PD-1, IL-10, etc). There was reduced NF-kB activation. Cancer-promoting immune cells (PD-1hiTim-3+ CTL, Tregs, M2-like macrophages, etc) occupied a minor spatial distribution, while the antitumor subtypes increased, corresponding to favorable survival. CONCLUSION H. pylori-evoked inflammatory responses disturb the TIME, causing a differential prognosis in de novo gDLBCL, which can be used to identify patients who could benefit from HPE and immunochemotherapy.
Collapse
Affiliation(s)
- Yuwei Deng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Wenjia Su
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junwen Zhu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Hongfei Ji
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaoping Zhou
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Jingshu Geng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Jiayu Zhu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
122
|
A Germinal Center-Associated Microenvironmental Signature Reflects Malignant Phenotype and Outcome of DLBCL. Blood Adv 2021; 6:2388-2402. [PMID: 34638128 PMCID: PMC9006269 DOI: 10.1182/bloodadvances.2021004618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/29/2021] [Indexed: 12/03/2022] Open
Abstract
The DLBCL microenvironment signature scoring system was established using nCounter-based profiling of GC-related microenvironmental genes. DMS scores stratified DLBCL patients with different prognosis independently of existing prognostic models.
Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell malignancy, with varying prognosis after the gold standard rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP). Several prognostic models have been established by focusing primarily on characteristics of lymphoma cells themselves, including cell-of-origin (COO), genomic alterations, and gene/protein expressions. However, the prognostic impact of the lymphoma microenvironment and its association with characteristics of lymphoma cells are not fully understood. Using the nCounter-based gene expression profiling of untreated DLBCL tissues, we assess the clinical impact of lymphoma microenvironment on the clinical outcomes and pathophysiological, molecular signatures in DLBCL. The presence of normal germinal center (GC)-microenvironmental cells, including follicular T cells, macrophage/dendritic cells, and stromal cells in lymphoma tissue indicates a positive therapeutic response. Our prognostic model, based on quantitation of transcripts from distinct GC-microenvironmental cell markers, clearly identified patients with graded prognosis independently of existing prognostic models. We observed increased incidences of genomic alterations and aberrant gene expression associated with poor prognosis in DLBCL tissues lacking GC-microenvironmental cells relative to those containing these cells. These data suggest that the loss of GC-associated microenvironmental signature dictates clinical outcomes of DLBCL patients reflecting the accumulation of “unfavorable” molecular signatures.
Collapse
|
123
|
Abstract
Diffuse large B cell lymphoma (DLBCL) is a markedly phenotypically heterogenous disease, frequently assessed using bulk genomic techniques that blur the intrinsic heterogeneity of each tumor. In this issue of Cancer Cell, Steen et al. have utilized a computational framework called EcoTyper to skillfully dissect bulk transcriptomic tumor profiles into different cell type components in an unsupervised manner.
Collapse
Affiliation(s)
- Dylan R McNally
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Ari Melnick
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA.
| |
Collapse
|
124
|
Steen CB, Luca BA, Esfahani MS, Azizi A, Sworder BJ, Nabet BY, Kurtz DM, Liu CL, Khameneh F, Advani RH, Natkunam Y, Myklebust JH, Diehn M, Gentles AJ, Newman AM, Alizadeh AA. The landscape of tumor cell states and ecosystems in diffuse large B cell lymphoma. Cancer Cell 2021; 39:1422-1437.e10. [PMID: 34597589 PMCID: PMC9205168 DOI: 10.1016/j.ccell.2021.08.011] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 06/24/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Biological heterogeneity in diffuse large B cell lymphoma (DLBCL) is partly driven by cell-of-origin subtypes and associated genomic lesions, but also by diverse cell types and cell states in the tumor microenvironment (TME). However, dissecting these cell states and their clinical relevance at scale remains challenging. Here, we implemented EcoTyper, a machine-learning framework integrating transcriptome deconvolution and single-cell RNA sequencing, to characterize clinically relevant DLBCL cell states and ecosystems. Using this approach, we identified five cell states of malignant B cells that vary in prognostic associations and differentiation status. We also identified striking variation in cell states for 12 other lineages comprising the TME and forming cell state interactions in stereotyped ecosystems. While cell-of-origin subtypes have distinct TME composition, DLBCL ecosystems capture clinical heterogeneity within existing subtypes and extend beyond cell-of-origin and genotypic classes. These results resolve the DLBCL microenvironment at systems-level resolution and identify opportunities for therapeutic targeting (https://ecotyper.stanford.edu/lymphoma).
Collapse
Affiliation(s)
- Chloé B Steen
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Bogdan A Luca
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mohammad S Esfahani
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | - Armon Azizi
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Brian J Sworder
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | - Barzin Y Nabet
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, CA 94305, USA
| | - David M Kurtz
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | - Chih Long Liu
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | - Farnaz Khameneh
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Ranjana H Advani
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University Medical Center, Stanford, CA 94305, USA
| | - June H Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B-cell malignancies, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University Medical Center, Stanford, CA 94305, USA
| | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Stanford Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA.
| | - Ash A Alizadeh
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
125
|
Garcia-Lacarte M, Grijalba SC, Melchor J, Arnaiz-Leché A, Roa S. The PD-1/PD-L1 Checkpoint in Normal Germinal Centers and Diffuse Large B-Cell Lymphomas. Cancers (Basel) 2021; 13:4683. [PMID: 34572910 PMCID: PMC8471895 DOI: 10.3390/cancers13184683] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/20/2022] Open
Abstract
Besides a recognized role of PD-1/PD-L1 checkpoint in anti-tumour immune evasion, there is accumulating evidence that PD-1/PD-L1 interactions between B and T cells also play an important role in normal germinal center (GC) reactions. Even when smaller in number, T follicular helper cells (TFH) and regulatory T (TFR) or B (Breg) cells are involved in positive selection of GC B cells and may result critical in the lymphoma microenvironment. Here, we discuss a role of PD-1/PD-L1 during tumour evolution in diffuse large B cell lymphoma (DLBCL), a paradigm of GC-derived lymphomagenesis. We depict a progression model, in two phases, where malignant B cells take advantage of positive selection signals derived from correct antigen-presentation and PD-1/PD-L1 inter-cellular crosstalks to survive and initiate tumour expansion. Later, a constant pressure for the accumulation of genetic/epigenetic alterations facilitates that DLBCL cells exhibit higher PD-L1 levels and capacity to secrete IL-10, resembling Breg-like features. As a result, a complex immunosuppressive microenvironment is established where DLBCL cells sustain proliferation and survival by impairing regulatory control of TFR cells and limiting IL-21-mediated anti-tumour functions of TFH cells and maximize the use of PD-1/PD-L1 signaling to escape from CD8+ cytotoxic activity. Integration of these molecular and cellular addictions into a framework may contribute to the better understanding of the lymphoma microenvironment and contribute to the rationale for novel PD-1/PD-L1-based combinational immunotherapies in DLBCL.
Collapse
Affiliation(s)
- Marcos Garcia-Lacarte
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
- Hemato-Oncology Program, Cima University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Sara C. Grijalba
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
| | - Javier Melchor
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
- Hemato-Oncology Program, Cima University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Adrián Arnaiz-Leché
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
| | - Sergio Roa
- Department of Biochemistry and Genetics, University of Navarra, 31008 Pamplona, Spain; (M.G.-L.); (S.C.G.); (J.M.); (A.A.-L.)
- Hemato-Oncology Program, Cima University of Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Network Center for Biomedical Research in Cancer—Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|
126
|
Croci GA, Au-Yeung RKH, Reinke S, Staiger AM, Koch K, Oschlies I, Richter J, Poeschel V, Held G, Loeffler M, Trümper L, Rosenwald A, Ott G, Spang R, Altmann B, Ziepert M, Klapper W. SPARC-positive macrophages are the superior prognostic factor in the microenvironment of diffuse large B-cell lymphoma and independent of MYC rearrangement and double-/triple-hit status. Ann Oncol 2021; 32:1400-1409. [PMID: 34438040 DOI: 10.1016/j.annonc.2021.08.1991] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease with respect to outcome. Features of the tumor microenvironment (TME) are associated with prognosis when assessed by gene expression profiling. However, it is uncertain whether assessment of the microenvironment can add prognostic information to the most relevant and clinically well-established molecular subgroups when analyzed by immunohistochemistry (IHC). PATIENTS AND METHODS We carried out a histopathologic analysis of biomarkers related to TME in a very large cohort (n = 455) of DLBCL treated in prospective trials and correlated with clinicopathologic and molecular data, including chromosomal rearrangements and gene expression profiles for cell-of-origin and TME. RESULTS The content of PD1+, FoxP3+ and CD8+, as well as vessel density, was not associated with outcome. However, we found a low content of CD68+ macrophages to be associated with inferior progression-free survival (PFS) and overall survival (OS; P = 0.023 and 0.040, respectively) at both univariable and multivariable analyses, adjusted for the factors of the International Prognostic Index (IPI), MYC break and BCL2/MYC and BCL6/MYC double-hit status. The subgroup of PDL1+ macrophages was not associated with survival. Instead, secreted protein acidic and cysteine rich (SPARC)-positive macrophages were identified as the subtype of macrophages most associated with survival. SPARC-positive macrophages and stromal cells directly correlated with favorable PFS and OS (both, P[log rank] <0.001, P[trend] < 0.001). The association of SPARC with prognosis was independent of the factors of the IPI, MYC double-/triple-hit status, Bcl2/c-myc double expression, cell-of-origin subtype and a recently published gene expression signature [lymphoma-associated macrophage interaction signature (LAMIS)]. CONCLUSIONS SPARC expression in the TME detected by a single IHC staining with fair-to-good interobserver reproducibility is a powerful prognostic parameter. Thus SPARC expression is a strong candidate for risk assessment in DLBCL in daily practice.
Collapse
Affiliation(s)
- G A Croci
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy; Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - R K H Au-Yeung
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department of Pathology, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - S Reinke
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - A M Staiger
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Stuttgart, Germany; Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Tübingen, Germany
| | - K Koch
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - I Oschlies
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - J Richter
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - V Poeschel
- Department of Internal Medicine 1 (Oncology, Hematology, Clinical Immunology, and Rheumatology), Saarland University Medical School, Homburg/Saar, Germany
| | - G Held
- DSHNHL Studiensekretariat, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - M Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - L Trümper
- Department of Hematology and Oncology, Georg-August Universität, Göttingen, Germany
| | - A Rosenwald
- Institute of Pathology, Universität Würzburg and Comprehensive Cancer Center Mainfranken (CCCMF), Würzburg, Germany
| | - G Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus, Stuttgart, Germany; Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Tübingen, Germany
| | - R Spang
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - B Altmann
- DSHNHL Studiensekretariat, Universitätsklinikum des Saarlandes, Homburg, Germany
| | - M Ziepert
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - W Klapper
- Institute of Pathology, Hematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
127
|
Mossadegh-Keller N, Brisou G, Beyou A, Nadel B, Roulland S. Human B Lymphomas Reveal Their Secrets Through Genetic Mouse Models. Front Immunol 2021; 12:683597. [PMID: 34335584 PMCID: PMC8323519 DOI: 10.3389/fimmu.2021.683597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/12/2021] [Indexed: 12/18/2022] Open
Abstract
Lymphomas are cancers deriving from lymphocytes, arising preferentially in secondary lymphoid organs, and represent the 6th cancer worldwide and the most frequent blood cancer. The majority of B cell Non-Hodgkin lymphomas (B-NHL) develop from germinal center (GC) experienced mature B cells. GCs are transient structures that form in lymphoid organs in response to antigen exposure of naive B cells, and where B cell receptor (BCR) affinity maturation occurs to promote B cell differentiation into memory B and plasma cells producing high-affinity antibodies. Genomic instability associated with the somatic hypermutation (SHM) and class-switch recombination (CSR) processes during GC transit enhance susceptibility to malignant transformation. Most B cell differentiation steps in the GC are at the origin of frequent B cell malignant entities, namely Follicular Lymphoma (FL) and GCB diffuse large B cell lymphomas (GCB-DLBCL). Over the past decade, large sequencing efforts have provided a great boost in the identification of candidate oncogenes and tumor suppressors involved in FL and DLBCL oncogenesis. Mouse models have been instrumental to accurately mimic in vivo lymphoma-specific mutations and interrogate their normal function in the GC context and their oncogenic function leading to lymphoma onset. The limited access of biopsies during the initiating steps of the disease, the cellular and (epi)genetic heterogeneity of individual tumors across and within patients linked to perturbed dynamics of GC ecosystems make the development of genetically engineered mouse models crucial to decipher lymphomagenesis and disease progression and eventually to test the effects of novel targeted therapies. In this review, we provide an overview of some of the important genetically engineered mouse models that have been developed to recapitulate lymphoma-associated (epi)genetic alterations of two frequent GC-derived lymphoma entities: FL and GCB-DLCBL and describe how those mouse models have improved our knowledge of the molecular processes supporting GC B cell transformation.
Collapse
Affiliation(s)
| | - Gabriel Brisou
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Department of Hematology, Institut Paoli-Calmettes, Marseille, France
| | - Alicia Beyou
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Bertrand Nadel
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|
128
|
Park Y, Heider D, Hauschild AC. Integrative Analysis of Next-Generation Sequencing for Next-Generation Cancer Research toward Artificial Intelligence. Cancers (Basel) 2021; 13:3148. [PMID: 34202427 PMCID: PMC8269018 DOI: 10.3390/cancers13133148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
The rapid improvement of next-generation sequencing (NGS) technologies and their application in large-scale cohorts in cancer research led to common challenges of big data. It opened a new research area incorporating systems biology and machine learning. As large-scale NGS data accumulated, sophisticated data analysis methods became indispensable. In addition, NGS data have been integrated with systems biology to build better predictive models to determine the characteristics of tumors and tumor subtypes. Therefore, various machine learning algorithms were introduced to identify underlying biological mechanisms. In this work, we review novel technologies developed for NGS data analysis, and we describe how these computational methodologies integrate systems biology and omics data. Subsequently, we discuss how deep neural networks outperform other approaches, the potential of graph neural networks (GNN) in systems biology, and the limitations in NGS biomedical research. To reflect on the various challenges and corresponding computational solutions, we will discuss the following three topics: (i) molecular characteristics, (ii) tumor heterogeneity, and (iii) drug discovery. We conclude that machine learning and network-based approaches can add valuable insights and build highly accurate models. However, a well-informed choice of learning algorithm and biological network information is crucial for the success of each specific research question.
Collapse
Affiliation(s)
- Youngjun Park
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
| | - Dominik Heider
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
| | - Anne-Christin Hauschild
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
- Department of Medical Informatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
129
|
An Overview on Diffuse Large B-Cell Lymphoma Models: Towards a Functional Genomics Approach. Cancers (Basel) 2021; 13:cancers13122893. [PMID: 34207773 PMCID: PMC8226720 DOI: 10.3390/cancers13122893] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Lymphoma research is a paradigm of integrating basic and applied research within the fields of molecular marker-based diagnosis and therapy. In recent years, major advances in next-generation sequencing have substantially improved the understanding of the genomics underlying diffuse large B-cell lymphoma (DLBCL), the most frequent type of B-cell lymphoma. This review addresses the various approaches that have helped unveil the biology and intricate alterations in this pathology, from cell lines to more sophisticated last-generation experimental models, such as organoids. We also provide an overview of the most recent findings in the field, their potential relevance for designing targeted therapies and the corresponding applicability to personalized medicine. Abstract Lymphoma research is a paradigm of the integration of basic and clinical research within the fields of diagnosis and therapy. Clinical, phenotypic, and genetic data are currently used to predict which patients could benefit from standard treatment. However, alternative therapies for patients at higher risk from refractoriness or relapse are usually empirically proposed, based on trial and error, without considering the genetic complexity of aggressive B-cell lymphomas. This is primarily due to the intricate mosaic of genetic and epigenetic alterations in lymphomas, which are an obstacle to the prediction of which drug will work for any given patient. Matching a patient’s genes to drug sensitivity by directly testing live tissues comprises the “precision medicine” concept. However, in the case of lymphomas, this concept should be expanded beyond genomics, eventually providing better treatment options for patients in need of alternative therapeutic approaches. We provide an overview of the most recent findings in diffuse large B-cell lymphomas genomics, from the classic functional models used to study tumor biology and the response to experimental treatments using cell lines and mouse models, to the most recent approaches with spheroid/organoid models. We also discuss their potential relevance and applicability to daily clinical practice.
Collapse
|
130
|
Papageorgiou SG, Thomopoulos TP, Katagas I, Bouchla A, Pappa V. Prognostic molecular biomarkers in diffuse large B-cell lymphoma in the rituximab era and their therapeutic implications. Ther Adv Hematol 2021; 12:20406207211013987. [PMID: 34104369 PMCID: PMC8150462 DOI: 10.1177/20406207211013987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) represents a group of tumors characterized by substantial heterogeneity in terms of their pathological and biological features, a causal factor of their varied clinical outcome. This variation has persisted despite the implementation of rituximab in treatment regimens over the last 20 years. In this context, prognostic biomarkers are of great importance in order to identify high-risk patients that might benefit from treatment intensification or the introduction of novel therapeutic agents. Herein, we review current knowledge on specific immunohistochemical or genetic biomarkers that might be useful in clinical practice. Gene-expression profiling is a tool of special consideration in this effort, as it has enriched our understanding of DLBCL biology and has allowed for the classification of DLBCL by cell-of-origin as well as by more elaborate molecular signatures based on distinct gene-expression profiles. These subgroups might outperform individual biomarkers in terms of prognostication; however, their use in clinical practice is still limited. Moreover, the underappreciated role of the tumor microenvironment in DLBCL prognosis is discussed in terms of prognostic gene-expression signatures, as well as in terms of individual biomarkers of prognostic significance. Finally, the efficacy of novel therapeutic agents for the treatment of DLBCL patients are discussed and an evidence-based therapeutic approach by specific genetic subgroup is suggested.
Collapse
Affiliation(s)
- Sotirios G. Papageorgiou
- Second Department of Internal Medicine and Research Unit, University General Hospital ‘Attikon’, 1 Rimini Street, Haidari, Athens 12462, Greece
| | - Thomas P. Thomopoulos
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| | - Ioannis Katagas
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| | - Anthi Bouchla
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| | - Vassiliki Pappa
- Second Department of Internal Medicine and Research Unit, Hematology Unit, University General Hospital, ‘Attikon’, Haidari, Athens, Greece
| |
Collapse
|
131
|
Pileri SA, Mazzara S, Derenzini E. Plasmablastic lymphoma: one or more tumours? Haematologica 2021; 106:2542-2543. [PMID: 33951892 PMCID: PMC8485687 DOI: 10.3324/haematol.2021.278841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Indexed: 11/09/2022] Open
Affiliation(s)
- Stefano A Pileri
- Divisions of Haematopathology, Haematology Programme, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan (Italy).
| | - Saveria Mazzara
- Divisions of Haematopathology, Haematology Programme, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan (Italy)
| | - Enrico Derenzini
- Divisions of Haemato-oncology, Haematology Programme, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan (Italy)
| |
Collapse
|
132
|
Pileri SA, Tripodo C, Melle F, Motta G, Tabanelli V, Fiori S, Vegliante MC, Mazzara S, Ciavarella S, Derenzini E. Predictive and Prognostic Molecular Factors in Diffuse Large B-Cell Lymphomas. Cells 2021; 10:cells10030675. [PMID: 33803671 PMCID: PMC8003012 DOI: 10.3390/cells10030675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the commonest form of lymphoid malignancy, with a prevalence of about 40% worldwide. Its classification encompasses a common form, also termed as “not otherwise specified” (NOS), and a series of variants, which are rare and at least in part related to viral agents. Over the last two decades, DLBCL-NOS, which accounts for more than 80% of the neoplasms included in the DLBCL chapter, has been the object of an increasing number of molecular studies which have led to the identification of prognostic/predictive factors that are increasingly entering daily practice. In this review, the main achievements obtained by gene expression profiling (with respect to both neoplastic cells and the microenvironment) and next-generation sequencing will be discussed and compared. Only the amalgamation of molecular attributes will lead to the achievement of the long-term goal of using tailored therapies and possibly chemotherapy-free protocols capable of curing most (if not all) patients with minimal or no toxic effects.
Collapse
Affiliation(s)
- Stefano A. Pileri
- Division of Haematopathology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (F.M.); (G.M.); (V.T.); (S.F.); (S.M.)
- Correspondence: or
| | - Claudio Tripodo
- Tumor Immunology Unit, University of Palermo, 90133 Palermo, Italy;
- Tumor and Microenvironment Histopathology Unit, IFOM, the FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Federica Melle
- Division of Haematopathology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (F.M.); (G.M.); (V.T.); (S.F.); (S.M.)
| | - Giovanna Motta
- Division of Haematopathology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (F.M.); (G.M.); (V.T.); (S.F.); (S.M.)
| | - Valentina Tabanelli
- Division of Haematopathology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (F.M.); (G.M.); (V.T.); (S.F.); (S.M.)
| | - Stefano Fiori
- Division of Haematopathology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (F.M.); (G.M.); (V.T.); (S.F.); (S.M.)
| | - Maria Carmela Vegliante
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori ‘Giovanni Paolo II’, Viale Flacco 65, 70124 Bari, Italy; (M.C.V.); (S.C.)
| | - Saveria Mazzara
- Division of Haematopathology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy; (F.M.); (G.M.); (V.T.); (S.F.); (S.M.)
| | - Sabino Ciavarella
- Hematology and Cell Therapy Unit, IRCCS-Istituto Tumori ‘Giovanni Paolo II’, Viale Flacco 65, 70124 Bari, Italy; (M.C.V.); (S.C.)
| | - Enrico Derenzini
- Division of Haemato-Oncology, European Institute of Oncology, IEO IRCCS, Via Ripamonti 435, 20141 Milan, Italy;
- Department of Health Sciences, University of Milan, Via di Rudinì 8, 20146 Milan, Italy
| |
Collapse
|