1
|
Ohlstrom D, Walker ZJ, Pandey A, Davis LN, Engel KL, Pan Z, Forsberg PA, Mark TM, Gillen AE, Sherbenou DW. Single-Cell RNA Sequencing before and after Light Chain Escape Reveals Intrapatient Multiple Myeloma Subpopulations with Divergent Osteolytic Gene Expression. CANCER RESEARCH COMMUNICATIONS 2025; 5:106-118. [PMID: 39699274 PMCID: PMC11737298 DOI: 10.1158/2767-9764.crc-24-0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/09/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
SIGNIFICANCE scRNA-seq was used to study a patient with high-risk multiple myeloma featuring LCE. LCE was rooted in a transcriptomic subpopulation that corresponded to a genetic subclone and established novel links between LCE and LAMP5 overexpression to osteolysis and prognosis, validated in RNA-seq databases.
Collapse
Affiliation(s)
- Denis Ohlstrom
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Biomedical Sciences and Biotechnology, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zachary J. Walker
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Abhishek Pandey
- Hematology-Oncology Fellowship Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lorraine N. Davis
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Krysta L. Engel
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Zenggang Pan
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter A. Forsberg
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Tomer M. Mark
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Austin E. Gillen
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel W. Sherbenou
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
2
|
Zhang F, Zou M, Bai C, Zhu M. Prognostic signature based on S100 calcium-binding protein family members for lung adenocarcinoma and its clinical significance. Comput Methods Biomech Biomed Engin 2024:1-17. [PMID: 39012268 DOI: 10.1080/10255842.2024.2376668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
The S100 family proteins (S100s) participate in multiple stages of tumorigenesis and are considered to have potential value as biomarkers for detecting and predicting various cancers. But the role of S100s in lung adenocarcinoma (LUAD) prognosis is elusive. Transcriptional data of LUAD patients were retrieved from TCGA, and relevant literature was extensively reviewed to collect S100 genes. Differential gene expression analysis was performed on the LUAD data, followed by intersection analysis between the differentially expressed genes (DEGs) and S100 genes. Unsupervised consensus clustering analysis identified two clusters. Significant variations in overall survival between the two clusters were shown by Kaplan-Meier analysis. DEGs between the two clusters were analyzed using Lasso regression and univariate/multivariate Cox regression analysis, leading to construction of an 11-gene prognostic signature. The signature exhibited stable and accurate predictive capability in TCGA and GEO datasets. Subsequently, we observed distinct immune cell infiltration, immunotherapy response, and tumor mutation characteristics in high and low-risk groups. Finally, small molecular compounds targeting prognostic genes were screened using CellMiner database, and molecular docking confirmed the binding of AMG-176, Estramustine, and TAK-632 with prognostic genes. In conclusion, we generated a prognostic signature with robust and reliable predictive ability, which may provide guidance for prognosis and treatment of LUAD.
Collapse
Affiliation(s)
- Fengshun Zhang
- Department of Pathology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Mi Zou
- Respiratory Department, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunsheng Bai
- Academician Expert Workstation of Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
- Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
| | - Mengjiao Zhu
- Academician Expert Workstation of Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
- Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
| |
Collapse
|
3
|
Boyle EM, Blaney P, Stoeckle JH, Wang Y, Ghamlouch H, Gagler D, Braunstein M, Williams L, Tenenbaum A, Siegel A, Chen X, Varma G, Avigan J, Li A, Jinsi M, Kaminetzsky D, Arbini A, Montes L, Corre J, Rustad EH, Landgren O, Maura F, Walker BA, Bauer M, Bruno B, Tsirigos A, Davies FE, Morgan GJ. Multiomic Mapping of Acquired Chromosome 1 Copy-Number and Structural Variants to Identify Therapeutic Vulnerabilities in Multiple Myeloma. Clin Cancer Res 2023; 29:3901-3913. [PMID: 37449980 PMCID: PMC12166981 DOI: 10.1158/1078-0432.ccr-22-3209] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/27/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE Chromosome 1 (chr1) copy-number abnormalities (CNA) and structural variants (SV) are frequent in newly diagnosed multiple myeloma (NDMM) and are associated with a heterogeneous impact on outcomes, the drivers of which are largely unknown. EXPERIMENTAL DESIGN A multiomic approach comprising CRISPR, gene mapping of CNAs and SVs, methylation, expression, and mutational analysis was used to document the extent of chr1 molecular variants and their impact on pathway utilization. RESULTS We identified two distinct groups of gain(1q): focal gains associated with limited gene-expression changes and a neutral prognosis, and whole-arm gains, which are associated with substantial gene-expression changes, complex genetics, and an adverse prognosis. CRISPR identified a number of dependencies on chr1 but only limited variants associated with acquired CNAs. We identified seven regions of deletion, nine of gain, three of chromothripsis (CT), and two of templated insertion (TI), which contain a number of potential drivers. An additional mechanism involving hypomethylation of genes at 1q may contribute to the aberrant gene expression of a number of genes. Expression changes associated with whole-arm gains were substantial and gene set enrichment analysis identified metabolic processes, apoptotic resistance, signaling via the MAPK pathway, and upregulation of transcription factors as being key drivers of the adverse prognosis associated with these variants. CONCLUSIONS Multiple layers of genetic complexity impact the phenotype associated with CNAs on chr1 to generate its associated clinical phenotype. Whole-arm gains of 1q are the critically important prognostic group that deregulate multiple pathways, which may offer therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Eileen M Boyle
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Patrick Blaney
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, New York
| | - James H Stoeckle
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Yubao Wang
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Hussein Ghamlouch
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Dylan Gagler
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, New York
| | - Marc Braunstein
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Louis Williams
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
- Myeloma Group, Cleveland Clinic Foundation, Taussig Cancer Center, Cleveland, Ohio
| | - Avital Tenenbaum
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Ariel Siegel
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Xiaoyi Chen
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Gaurav Varma
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Jason Avigan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Alexander Li
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Monica Jinsi
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - David Kaminetzsky
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Arnaldo Arbini
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | | | - Jill Corre
- Unit for Genomics in Myeloma, Institut Universitaire du Cancer de Toulouse-Oncopole, University Hospital, Toulouse; Centre de Recherche en Cancérologie de Toulouse, Institut National de la Santé et de la Recherche Médicale U1037, Toulouse, France
| | - Even H Rustad
- Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| | - Ola Landgren
- Myeloma Service, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Francesco Maura
- Myeloma Service, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Brian A Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Department of Hematology Oncology, Indiana University, Indianapolis, Indiana
| | - Michael Bauer
- Department of Biomedical Informatics (DBMI), UAMS, Little-Rock, Arkansas
| | - Benedetto Bruno
- Department of Hematology, Azienda Ospedaliera Citta della Salute e della Scienza di Torino, Piemonte, Italy
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, NYU Langone Medical Center, New York, New York
| | - Faith E Davies
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| | - Gareth J Morgan
- Myeloma Research Program, Perlmutter Cancer Center, NYU Langone Medical Center, New York, New York
| |
Collapse
|
4
|
Forster S, Radpour R, Ochsenbein AF. Molecular and immunological mechanisms of clonal evolution in multiple myeloma. Front Immunol 2023; 14:1243997. [PMID: 37744361 PMCID: PMC10516567 DOI: 10.3389/fimmu.2023.1243997] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by the proliferation of clonal plasma cells in the bone marrow (BM). It is known that early genetic mutations in post-germinal center B/plasma cells are the cause of myelomagenesis. The acquisition of additional chromosomal abnormalities and distinct mutations further promote the outgrowth of malignant plasma cell populations that are resistant to conventional treatments, finally resulting in relapsed and therapy-refractory terminal stages of MM. In addition, myeloma cells are supported by autocrine signaling pathways and the tumor microenvironment (TME), which consists of diverse cell types such as stromal cells, immune cells, and components of the extracellular matrix. The TME provides essential signals and stimuli that induce proliferation and/or prevent apoptosis. In particular, the molecular pathways by which MM cells interact with the TME are crucial for the development of MM. To generate successful therapies and prevent MM recurrence, a thorough understanding of the molecular mechanisms that drive MM progression and therapy resistance is essential. In this review, we summarize key mechanisms that promote myelomagenesis and drive the clonal expansion in the course of MM progression such as autocrine signaling cascades, as well as direct and indirect interactions between the TME and malignant plasma cells. In addition, we highlight drug-resistance mechanisms and emerging therapies that are currently tested in clinical trials to overcome therapy-refractory MM stages.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F. Ochsenbein
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Chen X, Chen S, Thomson M. Minimal gene set discovery in single-cell mRNA-seq datasets with ActiveSVM. NATURE COMPUTATIONAL SCIENCE 2022; 2:387-398. [PMID: 38177588 PMCID: PMC10766518 DOI: 10.1038/s43588-022-00263-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 05/17/2022] [Indexed: 01/06/2024]
Abstract
Sequencing costs currently prohibit the application of single-cell mRNA-seq to many biological and clinical analyses. Targeted single-cell mRNA-sequencing reduces sequencing costs by profiling reduced gene sets that capture biological information with a minimal number of genes. Here we introduce an active learning method that identifies minimal but highly informative gene sets that enable the identification of cell types, physiological states and genetic perturbations in single-cell data using a small number of genes. Our active feature selection procedure generates minimal gene sets from single-cell data by employing an active support vector machine (ActiveSVM) classifier. We demonstrate that ActiveSVM feature selection identifies gene sets that enable ~90% cell-type classification accuracy across, for example, cell atlas and disease-characterization datasets. The discovery of small but highly informative gene sets should enable reductions in the number of measurements necessary for application of single-cell mRNA-seq to clinical tests, therapeutic discovery and genetic screens.
Collapse
Affiliation(s)
- Xiaoqiao Chen
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, California, USA
| | - Sisi Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
- Beckman Institute Single-cell Profiling and Engineering Center, Pasadena, California, USA
| | - Matt Thomson
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, California, USA.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA.
- Beckman Institute Single-cell Profiling and Engineering Center, Pasadena, California, USA.
| |
Collapse
|