51
|
Sant P, Rippe K, Mallm JP. Approaches for single-cell RNA sequencing across tissues and cell types. Transcription 2023; 14:127-145. [PMID: 37062951 PMCID: PMC10807473 DOI: 10.1080/21541264.2023.2200721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
Single-cell sequencing of RNA (scRNA-seq) has advanced our understanding of cellular heterogeneity and signaling in developmental biology and disease. A large number of complementary assays have been developed to profile transcriptomes of individual cells, also in combination with other readouts, such as chromatin accessibility or antibody-based analysis of protein surface markers. As scRNA-seq technologies are advancing fast, it is challenging to establish robust workflows and up-to-date protocols that are best suited to address the large range of research questions. Here, we review scRNA-seq techniques from mRNA end-counting to total RNA in relation to their specific features and outline the necessary sample preparation steps and quality control measures. Based on our experience in dealing with the continuously growing portfolio from the perspective of a central single-cell facility, we aim to provide guidance on how workflows can be best automatized and share our experience in coping with the continuous expansion of scRNA-seq techniques.
Collapse
Affiliation(s)
- Pooja Sant
- Single-cell Open Lab, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| | - Karsten Rippe
- Division Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| | - Jan-Philipp Mallm
- Single-cell Open Lab, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| |
Collapse
|
52
|
Chen M, Jiang J, Hou J. Single-cell technologies in multiple myeloma: new insights into disease pathogenesis and translational implications. Biomark Res 2023; 11:55. [PMID: 37259170 PMCID: PMC10234006 DOI: 10.1186/s40364-023-00502-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by clonal proliferation of plasma cells. Although therapeutic advances have been made to improve clinical outcomes and to prolong patients' survival in the past two decades, MM remains largely incurable. Single-cell sequencing (SCS) is a powerful method to dissect the cellular and molecular landscape at single-cell resolution, instead of providing averaged results. The application of single-cell technologies promises to address outstanding questions in myeloma biology and has revolutionized our understanding of the inter- and intra-tumor heterogeneity, tumor microenvironment, and mechanisms of therapeutic resistance in MM. In this review, we summarize the recently developed SCS methodologies and latest MM research progress achieved by single-cell profiling, including information regarding the cancer and immune cell landscapes, tumor heterogeneities, underlying mechanisms and biomarkers associated with therapeutic response and resistance. We also discuss future directions of applying transformative SCS approaches with contribution to clinical translation.
Collapse
Affiliation(s)
- Mengping Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jinxing Jiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
53
|
Suzuki K, Yano S. Treatment Strategy for Ultra-High-Risk Multiple Myelomas with Chromosomal Aberrations Considering Minimal Residual Disease Status and Bone Marrow Microenvironment. Cancers (Basel) 2023; 15:cancers15092418. [PMID: 37173885 PMCID: PMC10177433 DOI: 10.3390/cancers15092418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the development of anti-myeloma therapeutics, such as proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies, and autologous stem cell transplantation (ASCT), multiple myeloma remains incurable. A trial treatment combining four drugs-daratumumab, carfilzomib, lenalidomide, and dexamethasone-followed by ASCT frequently results in minimal residual disease (MRD) negativity and prevents progressive disease in patients with standard- and high-risk cytogenetics; however, it is insufficient to overcome the poor outcomes in patients with ultra-high-risk chromosomal aberration (UHRCA). In fact, MRD status in autografts can predict clinical outcomes after ASCT. Therefore, the current treatment strategy might be insufficient to overcome the negative impact of UHRCA in patients with MRD positivity after the four-drug induction therapy. High-risk myeloma cells lead to poor clinical outcomes not only by aggressive myeloma behavior but also via the generation of a poor bone marrow microenvironment. Meanwhile, the immune microenvironment effectively suppresses myeloma cells with a low frequency of high-risk cytogenetic abnormalities in early-stage myeloma compared to late-stage myeloma. Therefore, early intervention might be key to improving clinical outcomes in myeloma patients. The purpose of this review is to improve clinical outcomes in patients with UHRCA by considering MRD assessment results and improvement of the microenvironment.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
54
|
Wan Y, Chen M, Li X, Han X, Zhong L, Xiao F, Liu J, Xiang J, Jiang J, Chen X, Liu J, Li H, Li B, Huang H, Hou J. Single-cell RNA sequencing reveals XBP1-SLC38A2 axis as a metabolic regulator in cytotoxic T lymphocytes in multiple myeloma. Cancer Lett 2023; 562:216171. [PMID: 37054944 DOI: 10.1016/j.canlet.2023.216171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/15/2023]
Abstract
The mechanisms underlying the functional impairment and metabolic reprogramming of T lymphocytes in multiple myeloma (MM) have not been fully elucidated. In this study, single-cell RNA sequencing was used to compare gene expression profiles in T cells in bone marrow and peripheral blood of 10 newly diagnosed MM patients versus 3 healthy donors. Unbiased bioinformatics analysis revealed 9 cytotoxic T cell clusters. All 9 clusters in MM had higher expression of senescence markers (e.g., KLRG1 and CTSW) than the healthy control; some had higher expression of exhaustion-related markers (e.g., LAG3 and TNFRSF14). Pathway enrichment analyses showed downregulated amino acid metabolism and upregulated unfolded protein response (UPR) pathways, along with absent expression of glutamine transporter SLC38A2 and increased expression of UPR hallmark XBP1 in cytotoxic T cells in MM. In vitro studies revealed that XBP1 inhibited SLC38A2 by directly binding to its promoter, and silencing SLC38A2 resulted in decreased glutamine uptake and immune dysfunction of T cells. This study provided a landscape description of the immunosuppressive and metabolic features in T lymphocytes in MM, and suggested an important role of XBP1-SLC38A2 axis in T cell function.
Collapse
Affiliation(s)
- Yike Wan
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Mengping Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xin Li
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaofeng Han
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lu Zhong
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Fei Xiao
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jia Liu
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jing Xiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jinxing Jiang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaotong Chen
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hua Li
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Honghui Huang
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
55
|
Ren L, Xu B, Xu J, Li J, Jiang J, Ren Y, Liu P. A Machine Learning Model to Predict Survival and Therapeutic Responses in Multiple Myeloma. Int J Mol Sci 2023; 24:ijms24076683. [PMID: 37047654 PMCID: PMC10095137 DOI: 10.3390/ijms24076683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Multiple myeloma (MM) is a highly heterogeneous hematologic tumor. Ubiquitin proteasome pathways (UPP) play a vital role in its initiation and development. We used cox regression analysis and least absolute shrinkage and selector operation (LASSO) to select ubiquitin proteasome pathway associated genes (UPPGs) correlated with the overall survival (OS) of MM patients in a Gene Expression Omnibus (GEO) dataset, and we formed this into ubiquitin proteasome pathway risk score (UPPRS). The association between clinical outcomes and responses triggered by proteasome inhibitors (PIs) and UPPRS were evaluated. MMRF CoMMpass was used for validation. We applied machine learning algorithms to MM clinical and UPPRS in the whole cohort to make a prognostic nomogram. Single-cell data and vitro experiments were performed to unravel the mechanism and functions of UPPRS. UPPRS consisting of 9 genes showed a strong ability to predict OS in MM patients. Additionally, UPPRS can be used to sort out the patients who would gain more benefits from PIs. A machine learning model incorporating UPPRS and International Staging System (ISS) improved survival prediction in both datasets compared to the revisions of ISS. At the single-cell level, high-risk UPPRS myeloma cells exhibited increased cell adhesion. Targeted UPPGs effectively inhibited myeloma cells in vitro. The UPP genes risk score is a helpful tool for risk stratification in MM patients, particularly those treated with PIs.
Collapse
Affiliation(s)
- Liang Ren
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Bei Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jiadai Xu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jing Li
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jifeng Jiang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuhong Ren
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Peng Liu
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
56
|
Wang XX, Deng SZ, Wu LH, Liu QQ, Zheng G, Du K, Dou QY, Zheng J, Zhang HM. Cuproptosis-Mediated Patterns Characterized by Distinct Tumor Microenvironment and Predicted the Immunotherapy Response for Gastric Cancer. ACS OMEGA 2023; 8:10851-10862. [PMID: 37008098 PMCID: PMC10061503 DOI: 10.1021/acsomega.2c07052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/28/2023] [Indexed: 05/26/2023]
Abstract
Cuproptosis is a newly discovered programmed cell death process, and several cuproptosis-related genes have been reported to regulate cancer cell proliferation and progression. The association between cuproptosis and tumor microenvironment in gastric cancer (GC) remains unclear. This study aimed to explore multiomics characteristics of cuproptosis-related genes regulating tumor microenvironment and provide strategies for prognosis and prediction of immunotherapy response in GC patients. We collected 1401 GC patients from the TCGA and 5 GEO data sets and identified three different cuproptosis-mediated patterns, each of which shared a distinct tumor microenvironment and different overall survival. The GC patients with high cuproptosis levels were enriched in CD8+ T cells and had a better prognosis. Whereas, the low cuproptosis level patients were associated with inhibitory immune cell infiltration and had the worst prognosis. In addition, we constructed a 3-gene (AHCYL2, ANKRD6 and FDGFRB) cuproptosis-related prognosis signature (CuPS) via Lasso-Cox and multivariate Cox regression analysis. The GC patients in the low-CuPS subgroup had higher TMB levels, MSI-H fractions, and PD-L1 expression, which suggests a better immunotherapy response. Therefore, the CuPS might have the potential value for predicting prognosis and immunotherapy sensitivity in GC patients.
Collapse
Affiliation(s)
- Xiang-Xu Wang
- Department
of Clinical Oncology, Xijing Hospital, Fourth
Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Shi-Zhou Deng
- Department
of Clinical Oncology, Xijing Hospital, Fourth
Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Li-Hong Wu
- Xijing
986 Hospital Department, Fourth Military
Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Qing-Qing Liu
- Department
of Clinical Oncology, Xijing Hospital, Fourth
Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Gaozan Zheng
- Division
of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Kunli Du
- Division
of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Qiong-Yi Dou
- Department
of Clinical Oncology, Xijing Hospital, Fourth
Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Jianyong Zheng
- Division
of Digestive Surgery, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| | - Hong-Mei Zhang
- Department
of Clinical Oncology, Xijing Hospital, Fourth
Military Medical University, Xi’an, Shaanxi 710032, P. R. China
| |
Collapse
|
57
|
Russell BM, Avigan DE. Immune dysregulation in multiple myeloma: the current and future role of cell-based immunotherapy. Int J Hematol 2023; 117:652-659. [PMID: 36964840 PMCID: PMC10039687 DOI: 10.1007/s12185-023-03579-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 02/27/2023] [Accepted: 03/05/2023] [Indexed: 03/26/2023]
Abstract
Immune dysregulation is a hallmark of clinically active multiple myeloma (MM). Interactions between malignant clonal cells and immune cells within the bone marrow microenvironment are associated with the formation of a milieu favorable to tumor progression. IL-10, TGF-β and other immunoregulatory pathways are upregulated, promoting angiogenesis, tumor cell survival and inhibition of the native immune response. Transcriptomic evaluation of the bone marrow microenvironment reveals polarization of the T cell repertoire towards exhaustion and predominance of accessory cells with immunosuppressive qualities. These changes facilitate the immune escape of tumor cells and functional deficiencies that manifest as an increased risk of infection and a reduction in response to vaccinations. Immunotherapy with Chimeric Antigen Receptor (CAR) T cells and other cellular-based approaches have transformed outcomes for patients with advanced MM. Characterization of the immune milieu and identification of biomarkers predictive of treatment response are essential to increasing durability and allowing for the incorporation of novel strategies such as cancer vaccines. This paper will review the current use of cancer vaccines and CAR T cell therapy in MM as well as potential opportunities to expand and improve the application of these platforms.
Collapse
Affiliation(s)
- Brian M Russell
- Department of Medicine, Divisions of Hematology & Hematologic Malignancies, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02115, USA
| | - David E Avigan
- Department of Medicine, Divisions of Hematology & Hematologic Malignancies, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02115, USA.
| |
Collapse
|
58
|
Terpos E, Neri P, van de Donk NWCJ, D'Agostino M, Parekh S, Jagannath S, Ludwig H, Avigan DE, Dhodapkar MV, Raje NS. Immune Reconstitution and Vaccinations in Multiple Myeloma: A Report From the 19th International Myeloma Society Annual Workshop. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:413-419. [PMID: 37055346 DOI: 10.1016/j.clml.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
Given the significance of the immune system and the important role of therapies within the context of the immune system in plasma cell disorders, the International Myeloma Society annual workshop convened a session dedicated to this topic. A panel of experts covered various aspects of immune reconstitution and vaccination. The top oral presentations were highlighted and discussed. This is a report of the proceedings.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Paola Neri
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, Alberta, Canada; Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mattia D'Agostino
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Samir Parekh
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sundar Jagannath
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Heinz Ludwig
- Department of Medicine I, Center for Medical Oncology and Hematology with Outpatient Department and Palliative Care, Wilhelminen Cancer Research Institute, Vienna, Austria
| | - David E Avigan
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| | - Noopur S Raje
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA; Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
59
|
Friedrich MJ, Neri P, Kehl N, Michel J, Steiger S, Kilian M, Leblay N, Maity R, Sankowski R, Lee H, Barakat E, Ahn S, Weinhold N, Rippe K, Bunse L, Platten M, Goldschmidt H, Müller-Tidow C, Raab MS, Bahlis NJ. The pre-existing T cell landscape determines the response to bispecific T cell engagers in multiple myeloma patients. Cancer Cell 2023; 41:711-725.e6. [PMID: 36898378 DOI: 10.1016/j.ccell.2023.02.008] [Citation(s) in RCA: 93] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/02/2022] [Accepted: 02/08/2023] [Indexed: 03/11/2023]
Abstract
Bispecific T cell engagers (TCEs) have shown promise in the treatment of various cancers, but the immunological mechanism and molecular determinants of primary and acquired resistance to TCEs remain poorly understood. Here, we identify conserved behaviors of bone marrow-residing T cells in multiple myeloma patients undergoing BCMAxCD3 TCE therapy. We show that the immune repertoire reacts to TCE therapy with cell state-dependent clonal expansion and find evidence supporting the coupling of tumor recognition via major histocompatibility complex class I (MHC class I), exhaustion, and clinical response. We find the abundance of exhausted-like CD8+ T cell clones to be associated with clinical response failure, and we describe loss of target epitope and MHC class I as tumor-intrinsic adaptations to TCEs. These findings advance our understanding of the in vivo mechanism of TCE treatment in humans and provide the rationale for predictive immune-monitoring and conditioning of the immune repertoire to guide future immunotherapy in hematological malignancies.
Collapse
Affiliation(s)
- Mirco J Friedrich
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Paola Neri
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada; Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Niklas Kehl
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julius Michel
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Simon Steiger
- Division of Chromatin Networks, BioQuant Center & German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Michael Kilian
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Noémie Leblay
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Ranjan Maity
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Roman Sankowski
- Department of Neuropathology, Freiburg University Hospital, Freiburg, Germany
| | - Holly Lee
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada; Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Elie Barakat
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Sungwoo Ahn
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Niels Weinhold
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, BioQuant Center & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lukas Bunse
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Helmholtz Institute of Translational Oncology (HI-TRON), Mainz, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim Germany
| | - Hartmut Goldschmidt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Marc-Steffen Raab
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada; Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada.
| |
Collapse
|
60
|
Gao Z, Bai Y, Lin A, Jiang A, Zhou C, Cheng Q, Liu Z, Chen X, Zhang J, Luo P. Gamma delta T-cell-based immune checkpoint therapy: attractive candidate for antitumor treatment. Mol Cancer 2023; 22:31. [PMID: 36793048 PMCID: PMC9930367 DOI: 10.1186/s12943-023-01722-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
As a nontraditional T-cell subgroup, γδT cells have gained popularity in the field of immunotherapy in recent years. They have extraordinary antitumor potential and prospects for clinical application. Immune checkpoint inhibitors (ICIs), which are efficacious in tumor patients, have become pioneer drugs in the field of tumor immunotherapy since they were incorporated into clinical practice. In addition, γδT cells that have infiltrated into tumor tissues are found to be in a state of exhaustion or anergy, and there is upregulation of many immune checkpoints (ICs) on their surface, suggesting that γδT cells have a similar ability to respond to ICIs as traditional effector T cells. Studies have shown that targeting ICs can reverse the dysfunctional state of γδT cells in the tumor microenvironment (TME) and exert antitumor effects by improving γδT-cell proliferation and activation and enhancing cytotoxicity. Clarification of the functional state of γδT cells in the TME and the mechanisms underlying their interaction with ICs will solidify ICIs combined with γδT cells as a good treatment option.
Collapse
Affiliation(s)
- Zhifei Gao
- grid.284723.80000 0000 8877 7471The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong 510282 People’s Republic of China ,grid.284723.80000 0000 8877 7471The Second Clinical Medical School, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282 People’s Republic of China
| | - Yifeng Bai
- grid.54549.390000 0004 0369 4060The Department of Oncology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 611731 China
| | - Anqi Lin
- grid.284723.80000 0000 8877 7471The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong 510282 People’s Republic of China
| | - Aimin Jiang
- grid.73113.370000 0004 0369 1660The Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Chaozheng Zhou
- grid.284723.80000 0000 8877 7471The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong 510282 People’s Republic of China ,grid.284723.80000 0000 8877 7471The First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- grid.216417.70000 0001 0379 7164The Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan China ,grid.216417.70000 0001 0379 7164National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zaoqu Liu
- grid.412633.10000 0004 1799 0733The Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Xin Chen
- The Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jian Zhang
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, 510282, People's Republic of China.
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, Guangdong, 510282, People's Republic of China.
| |
Collapse
|
61
|
Laganà A. The genome of IMiD-refractory myeloma. Blood 2023; 141:560-561. [PMID: 36757732 DOI: 10.1182/blood.2022018461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
62
|
Chen M, Wan Y, Li X, Xiang J, Chen X, Jiang J, Han X, Zhong L, Xiao F, Liu J, Huang H, Li H, Liu J, Hou J. Dynamic single-cell RNA-seq analysis reveals distinct tumor program associated with microenvironmental remodeling and drug sensitivity in multiple myeloma. Cell Biosci 2023; 13:19. [PMID: 36717896 PMCID: PMC9887807 DOI: 10.1186/s13578-023-00971-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a hematological malignancy characterized by clonal proliferation of malignant plasma cells. Despite extensive research, molecular mechanisms in MM that drive drug sensitivity and clinic outcome remain elusive. RESULTS Single-cell RNA sequencing was applied to study tumor heterogeneity and molecular dynamics in 10 MM individuals before and after 2 cycles of bortezomib-cyclophosphamide-dexamethasone (VCD) treatment, with 3 healthy volunteers as controls. We identified that unfolded protein response and metabolic-related program were decreased, whereas stress-associated and immune reactive programs were increased after 2 cycles of VCD treatment. Interestingly, low expression of the immune reactive program by tumor cells was associated with unfavorable drug response and poor survival in MM, which probably due to downregulation of MHC class I mediated antigen presentation and immune surveillance, and upregulation of markers related to immune escape. Furthermore, combined with immune cells profiling, we uncovered a link between tumor intrinsic immune reactive program and immunosuppressive phenotype in microenvironment, evidenced by exhausted states and expression of checkpoint molecules and suppressive genes in T cells, NK cells and monocytes. Notably, expression of YBX1 was associated with downregulation of immune activation signaling in myeloma and reduced immune cells infiltration, thereby contributed to poor prognosis. CONCLUSIONS We dissected the tumor and immune reprogramming in MM during targeted therapy at the single-cell resolution, and identified a tumor program that integrated tumoral signaling and changes in immune microenvironment, which provided insights into understanding drug sensitivity in MM.
Collapse
Affiliation(s)
- Mengping Chen
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Yike Wan
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Xin Li
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Jing Xiang
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Xiaotong Chen
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Jinxing Jiang
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Xiaofeng Han
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Lu Zhong
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Fei Xiao
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Jia Liu
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Honghui Huang
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| | - Hua Li
- grid.16821.3c0000 0004 0368 8293Bio-ID Center, Shanghai Jiao Tong University School of Biomedical Engineering, Shanghai, 200240 China
| | - Junling Liu
- grid.16821.3c0000 0004 0368 8293Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jian Hou
- grid.16821.3c0000 0004 0368 8293Department of Hematology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127 China
| |
Collapse
|
63
|
Mizuguchi M, Okamoto Y, Yagi H, Kagawa K, Sekimoto E, Shibata H, Shigekiyo T, Ozaki S. Clinical relevance of high-risk cytogenetic abnormalities and the second revision of the International Staging System (R2-ISS) in patients with multiple myeloma in clinical practice. Int J Hematol 2023; 117:718-728. [PMID: 36692689 DOI: 10.1007/s12185-023-03541-x] [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: 11/08/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
High-risk cytogenetic abnormalities (HRCAs) are the most critical factor affecting prognosis in multiple myeloma (MM). However, the clinical significance of HRCAs in routine practice has not been fully elucidated. We retrospectively analyzed clinical features and outcome in 60 newly diagnosed MM patients with or without HRCAs including t(4;14), t(14;16), del(17p), and 1q gain/amplification. The median age was 71 years (range, 35-90). Abnormalities with t(4;14), t(14;16), del(17p), and 1q gain/amplification were found in 10, 1, 6, and 21/14 patients, respectively, and 10 patients had ≥ 2 HRCAs. Patients with HRCAs exhibited progressive clinical features such as anemia, high β2-microglobulin, and high LDH. Symptomatic relapse was more common in patients with HRCAs. The median progression-free survival (PFS) by number of HRCAs (0, 1, and ≥ 2) was 51.7, 21.4, and 26.1 months (p = 0.011), and the median overall survival (OS) was not reached, 60.7, and 46.8 months (p = 0.045), respectively. Multivariate analysis revealed that HRCAs were an independent factor for PFS. Accordingly, the second revision of International Staging System (R2-ISS), which incorporates HRCA scores, was more useful for prognostic stratification (p = 0.0023). These results suggest that presence of multiple HRCAs including 1q gain/amplification is associated with advanced stage and poor prognosis in clinical practice as well.
Collapse
Affiliation(s)
- Makiko Mizuguchi
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Yasunobu Okamoto
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Hikaru Yagi
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Kumiko Kagawa
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Etsuko Sekimoto
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan.,Department of Internal Medicine, Tenma Hospital, 1-5-1 Kuramoto, Tokushima, 770-0042, Japan
| | - Hironobu Shibata
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Toshio Shigekiyo
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan
| | - Shuji Ozaki
- Department of Hematology, Tokushima Prefecture Central Hospital, 1-10-3 Kuramoto, Tokushima, 770-8539, Japan.
| |
Collapse
|
64
|
Giannotta C, Autino F, Massaia M. Vγ9Vδ2 T-cell immunotherapy in blood cancers: ready for prime time? Front Immunol 2023; 14:1167443. [PMID: 37143664 PMCID: PMC10153673 DOI: 10.3389/fimmu.2023.1167443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
In the last years, the tumor microenvironment (TME) has emerged as a promising target for therapeutic interventions in cancer. Cancer cells are highly dependent on the TME to growth and evade the immune system. Three major cell subpopulations are facing each other in the TME: cancer cells, immune suppressor cells, and immune effector cells. These interactions are influenced by the tumor stroma which is composed of extracellular matrix, bystander cells, cytokines, and soluble factors. The TME can be very different depending on the tissue where cancer arises as in solid tumors vs blood cancers. Several studies have shown correlations between the clinical outcome and specific patterns of TME immune cell infiltration. In the recent years, a growing body of evidence suggests that unconventional T cells like natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ T cells are key players in the protumor or antitumor TME commitment in solid tumors and blood cancers. In this review, we will focus on γδ T cells, especially Vγ9Vδ2 T cells, to discuss their peculiarities, pros, and cons as potential targets of therapeutic interventions in blood cancers.
Collapse
Affiliation(s)
- Claudia Giannotta
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
| | - Federica Autino
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
| | - Massimo Massaia
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S. Croce e Carle, Cuneo, Italy
- *Correspondence: Massimo Massaia,
| |
Collapse
|
65
|
Lepore A, Kaci FN, Bubici C, Papa S. An Integrated Methodology to Quantify the Glycolytic Stress in Plasma Cell Myeloma in Response to Cytotoxic Drugs. Methods Mol Biol 2023; 2675:285-296. [PMID: 37258771 DOI: 10.1007/978-1-0716-3247-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy primarily localized within the bone marrow (BM). Myeloma plasma cells, like many other cancer cells, change their metabolism in response to internal and external stimuli. The main metabolic alterations of MM cells include deregulated glycolysis (commonly associated with enhanced uptake and utilization of glucose), lipid metabolism dysregulation, as well as deregulated mitochondrial respiration (commonly associated with the deregulated formation of reactive oxygen species). Over the past decade, the discovery of novel methodologies and the commercialization of sophisticated instrumentation and reagents have facilitated the detection of real-time changes in cellular bioenergetics. Of those, the Seahorse™ extracellular flux (XF) analyzer has been widely used to evaluate the glycolytic flux and mitochondrial respiration in many cell types. While adherent cell lines are easy to use with this technology, non-adherent suspension cells are more difficult to handle especially when their metabolic activities are being investigated in response to drug treatment. Here, we provide an integrated protocol that allows the detection of extracellular acidification rate (ECAR) of live myeloma plasma cells in response to chemotherapeutic drugs. Our optimized protocol consists of treating myeloma cells with cytotoxic drug of interest in a standard culture plate prior to the real-time analysis in the XF analyzer. Furthermore, we provide results of experiments in which the metabolic activities of myeloma cells in response to cytotoxic treatment were compared between the manufacturer's basic procedure and our optimized protocol. Our observations suggest that our integrated protocol can be used to achieve consistent, well-standardized results and thus it may have broad applications in studies focusing on the characterization of metabolic events in non-adherent suspension cells.
Collapse
Affiliation(s)
- Alessio Lepore
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Fatma Necmiye Kaci
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Concetta Bubici
- Department of Life Sciences, Center for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London, UK
| | - Salvatore Papa
- Leeds Institute of Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK.
| |
Collapse
|
66
|
Schmidt TM. High or low? Assessing disease risk in multiple myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:349-355. [PMID: 36485159 PMCID: PMC9820796 DOI: 10.1182/hematology.2022000347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Based upon the development of highly effective therapies such as immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies that target plasma cell biology, a dramatic improvement in overall survival has been observed for most patients with multiple myeloma (MM) over the past 2 decades. Although it is now commonplace for many patients with myeloma to live in excess of 10 years after diagnosis, unfortunately a large subset of patients continues to experience an aggressive disease course marked by substantial morbidity and early mortality. Many clinical biomarkers and staging systems in use today can help with prognostication, but accurate risk assessment can be difficult due to the presence of many different biomarkers with variable prognostic value. Furthermore, with the implementation of novel therapies and unprecedented rates of deep and durable responses, it is becoming apparent that risk assessment is best envisioned as a dynamic process that requires ongoing reevaluation. As risk and response-adapted approaches are becoming more commonplace, it is essential that clinicians understand the biological and prognostic implications of clinical, genomic, and response-based biomarkers in order to promote management strategies that will help improve both survival and quality of life for patients across the risk spectrum.
Collapse
Affiliation(s)
- Timothy Martin Schmidt
- Correspondence Timothy Martin Schmidt, University of Wisconsin, 600 Highland Avenue, K6/544 Clinical Sciences Center, MC5669, Madison, WI 53792; e-mail:
| |
Collapse
|
67
|
Molecular Crosstalk between Chromatin Remodeling and Tumor Microenvironment in Multiple Myeloma. Curr Oncol 2022; 29:9535-9549. [PMID: 36547163 PMCID: PMC9777166 DOI: 10.3390/curroncol29120749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a complex disease driven by numerous genetic and epigenetic alterations that are acquired over time. Despite recent progress in the understanding of MM pathobiology and the availability of innovative drugs, which have pronounced clinical outcome, this malignancy eventually progresses to a drug-resistant lethal stage and, thus, novel therapeutic drugs/models always play an important role in effective management of MM. Modulation of tumor microenvironment is one of the hallmarks of cancer biology, including MM, which affects the myeloma genomic architecture and disease progression subtly through chromatin modifications. The bone marrow niche has a prime role in progression, survival, and drug resistance of multiple myeloma cells. Therefore, it is important to develop means for targeting the ecosystem between multiple myeloma bone marrow microenvironment and chromatin remodeling. Extensive gene expression profile analysis has indeed provided the framework for new risk stratification of MM patients and identifying novel molecular targets and therapeutics. However, key tumor microenvironment factors/immune cells and their interactions with chromatin remodeling complex proteins that drive MM cell growth and progression remain grossly undefined.
Collapse
|
68
|
Lv J, Sun H, Gong L, Wei X, He Y, Yu Z, Liu L, Yi S, Sui W, Xu Y, Deng S, An G, Yao Z, Qiu L, Hao M. Aberrant metabolic processes promote the immunosuppressive microenvironment in multiple myeloma. Front Immunol 2022; 13:1077768. [PMID: 36532059 PMCID: PMC9748558 DOI: 10.3389/fimmu.2022.1077768] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Multiple myeloma (MM) is still an incurable plasma cell malignancy. The efficacy of immunotherapy on MM remains unsatisfactory, and the underlying molecular mechanisms still are not fully understood. Methods In this study, we delineated the dynamic features of immune cell in MM bone marrow (BM) along with elevated tumor cell infiltration by single-cell RNA sequencing (scRNA-seq), and investigated the underlying mechanisms on dysfunction of immune cells associated with myelomagenesis. Results We found that immune cells were activated in those patients with low infiltration of tumor cells, meanwhile suppressed with elevated infiltration of MM cells, which facilitated MM escaping from immune surveillance. Besides PD-1, abnormal expression of PIM kinases, KLRB1 and KLRC1 were involved in the defect of immune cells in MM patients. Importantly, we found aberrant metabolic processes were associated with the immunosuppressive microenvironment in MM patients. Disordered amino acid metabolism promoted the dysfunction of cytotoxicity CD8 T cells as well as lipid metabolism disorder was associated with the dysregulation of NK and DCs in MM. As metabolic checkpoints, PIM kinases would be potential effective strategies for MM immunotherapy. Discussion In summary, redressing the disordered metabolism should be the key points to get promising effects in immune-based therapies.
Collapse
Affiliation(s)
- Junqiang Lv
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hao Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Lixin Gong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaojing Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yi He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhen Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Lanting Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yan Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhui Deng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China
| | - Zhi Yao
- Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China,*Correspondence: Mu Hao, ; Lugui Qiu,
| | - Mu Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China,Tianjin Institutes of Health Science, Tianjin, China,*Correspondence: Mu Hao, ; Lugui Qiu,
| |
Collapse
|
69
|
Wang SSY, Chng WJ, Liu H, de Mel S. Tumor-Associated Macrophages and Related Myelomonocytic Cells in the Tumor Microenvironment of Multiple Myeloma. Cancers (Basel) 2022; 14:5654. [PMID: 36428745 PMCID: PMC9688291 DOI: 10.3390/cancers14225654] [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: 10/08/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM) is the second-most common hematologic malignancy and remains incurable despite potent plasma cell directed therapeutics. The tumor microenvironment (TME) is a key player in the pathogenesis and progression of MM and is an active focus of research with a view to targeting immune dysregulation. Tumor-associated macrophages (TAM), myeloid derived suppressor cells (MDSC), and dendritic cells (DC) are known to drive progression and treatment resistance in many cancers. They have also been shown to promote MM progression and immune suppression in vitro, and there is growing evidence of their impact on clinical outcomes. The heterogeneity and functional characteristics of myelomonocytic cells in MM are being unraveled through high-dimensional immune profiling techniques. We are also beginning to understand how they may affect and be modulated by current and future MM therapeutics. In this review, we provide an overview of the biology and clinical relevance of TAMs, MDSCs, and DCs in the MM TME. We also highlight key areas to be addressed in future research as well as our perspectives on how the myelomonocytic compartment of the TME may influence therapeutic strategies of the future.
Collapse
Affiliation(s)
- Samuel S. Y. Wang
- Department of Rheumatology, Allergy and Immunology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Wee Joo Chng
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
- Cancer Science Institute, National University of Singapore, 14 Medical Dr, #12-01 Centre for Translational Medicine, Singapore 117599, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
- Immunology Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Sanjay de Mel
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore 119228, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| |
Collapse
|
70
|
Kehl N, Kilian M, Michel J, Wagner TR, Uhrig S, Brobeil A, Sester LS, Blobner S, Steiger S, Hundemer M, Weinhold N, Rippe K, Fröhling S, Eichmüller SB, Bunse L, Müller-Tidow C, Goldschmidt H, Platten M, Raab MS, Friedrich MJ. IgE type multiple myeloma exhibits hypermutated phenotype and tumor reactive T cells. J Immunother Cancer 2022; 10:jitc-2022-005815. [PMID: 36252999 PMCID: PMC9577923 DOI: 10.1136/jitc-2022-005815] [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] [Accepted: 09/26/2022] [Indexed: 11/04/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy originating from malignant and clonally expanding plasma cells. MM can be molecularly stratified, and its clonal evolution deciphered based on the Ig heavy and light chains of the respective malignant plasma cell clone. Of all MM subtypes, IgE type MM accounts for only <0.1% of cases and is associated with an aggressive clinical course and consequentially dismal prognosis. In such malignancies, adoptive transfer of autologous lymphocytes specifically targeting presented (neo)epitopes encoded by either somatically mutated or specifically overexpressed genes has resulted in substantial objective clinical regressions even in relapsed/refractory disease. However, there are no data on the genetic and immunological characteristics of this rare and aggressive entity. Here, we comprehensively profiled IgE type kappa MM on a genomic and immune repertoire level by integrating DNA- and single-cell RNA sequencing and comparative profiling against non-IgE type MM samples. We demonstrate distinct pathophysiological mechanisms as well as novel opportunities for targeting IgE type MM. Our data further provides the rationale for patient-individualized neoepitope-targeting cell therapy in high tumor mutation burden MM.
Collapse
Affiliation(s)
- Niklas Kehl
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany,Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Kilian
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julius Michel
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany,Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tim R Wagner
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany,GMP and Cell Therapy Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Uhrig
- Molecular Precision Oncology Program, National Center for Tumor Diseases, Heidelberg, Germany
| | - Alexander Brobeil
- Department of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lilli-Sophie Sester
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sven Blobner
- Department of Translational Oncology, National Center of Tumor Diseases (NCT) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Simon Steiger
- Division of Chromatin Networks, BioQuant Center and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hundemer
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Niels Weinhold
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, BioQuant Center and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Oncology, National Center of Tumor Diseases (NCT) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP and Cell Therapy Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lukas Bunse
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hartmut Goldschmidt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany,National Center of Tumor Diseases (NCT) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany,Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany,National Center of Tumor Diseases (NCT) and German Cancer Consortium (DKTK), Heidelberg, Germany,DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Marc-Steffen Raab
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany,Clinical Cooperation Unit Molecular Hematology / Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirco J Friedrich
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
71
|
Masuda T, Haji S, Nakashima Y, Tsuda M, Kimura D, Takamatsu A, Iwahashi N, Umakoshi H, Shiratsuchi M, Kikutake C, Suyama M, Ohkawa Y, Ogawa Y. Identification of a drug-response gene in multiple myeloma through longitudinal single-cell transcriptome sequencing. iScience 2022; 25:104781. [PMID: 35992084 PMCID: PMC9386061 DOI: 10.1016/j.isci.2022.104781] [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: 01/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/06/2022] Open
Abstract
Despite recent therapeutic advances for multiple myeloma (MM), relapse is very common. Here, we conducted longitudinal single-cell transcriptome sequencing (scRNA-seq) of MM cells from a patient with relapsed MM, treated with multiple anti-myeloma drugs. We observed five subclusters of MM cells, which appeared and/or disappeared in response to the therapeutic pressure, and identified cluster 3 which emerged during lenalidomide treatment and disappeared after proteasome inhibitor (PI) treatment. Among the differentially expressed genes in cluster 3, we found a candidate drug-response gene; pellino E3 ubiquitin-protein ligase family member 2 (PELI2), which is responsible for PI-induced cell death in in vitro assay. Kaplan-Meier survival analysis of database revealed that higher expression of PELI2 is associated with a better prognosis. Our integrated strategy combining longitudinal scRNA-seq analysis, in vitro functional assay, and database analysis would facilitate the understanding of clonal dynamics of MM in response to anti-myeloma drugs and identification of drug-response genes.
Collapse
Affiliation(s)
- Toru Masuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shojiro Haji
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuhiro Nakashima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mariko Tsuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Daisaku Kimura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiko Takamatsu
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Norifusa Iwahashi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hironobu Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Motoaki Shiratsuchi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Hematology, Iizuka Hospital, Iizuka 820-8505, Japan
| | - Chie Kikutake
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
72
|
Gao X, Hong F, Hu Z, Zhang Z, Lei Y, Li X, Cheng T. ABC portal: a single-cell database and web server for blood cells. Nucleic Acids Res 2022; 51:D792-D804. [PMID: 35920330 PMCID: PMC9825444 DOI: 10.1093/nar/gkac646] [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: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 01/30/2023] Open
Abstract
ABC portal (http://abc.sklehabc.com) is a database and web portal containing 198 single-cell transcriptomic datasets of development, differentiation and disorder of blood/immune cells. All the datasets were re-annotated with a manually curated and unified single-cell reference, especially for the haematopoietic stem and progenitor cells. ABC portal provides web-based interactive analysis modules, especially a comprehensive cell-cell communication analysis and disease-related gene signature analysis. Importantly, ABC portal allows customized sample selection based on a combination of several metadata for downstream analysis and comparison analysis across datasets. ABC portal also allows users to select multiple cell types for analysis in the modules. Together, ABC portal provides an interactive interface of single-cell data exploration and re-analysis with customized analysis modules for the researchers and clinicians, and will facilitate understanding of haematopoiesis and blood/immune disorders.
Collapse
Affiliation(s)
- Xin Gao
- To whom correspondence should be addressed. Tel: +86 22 2390 9006; Fax: +86 22 2390 9006;
| | - Fang Hong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Zhenyu Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Zilong Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Yang Lei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Xiaoyun Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Tao Cheng
- Correspondence may also be addressed to Tao Cheng.
| |
Collapse
|
73
|
Li Z, Seehawer M, Polyak K. Untangling the web of intratumour heterogeneity. Nat Cell Biol 2022; 24:1192-1201. [PMID: 35941364 DOI: 10.1038/s41556-022-00969-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 06/27/2022] [Indexed: 02/06/2023]
Abstract
Intratumour heterogeneity (ITH) is a hallmark of cancer that drives tumour evolution and disease progression. Technological and computational advances have enabled us to assess ITH at unprecedented depths, yet this accumulating knowledge has not had a substantial clinical impact. This is in part due to a limited understanding of the functional relevance of ITH and the inadequacy of preclinical experimental models to reproduce it. Here, we discuss progress made in these areas and illuminate future directions.
Collapse
Affiliation(s)
- Zheqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Marco Seehawer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
74
|
Soekojo CY, Chng WJ. The Evolution Of Immune Dysfunction In Multiple Myeloma. Eur J Haematol 2022; 109:415-424. [PMID: 35880386 DOI: 10.1111/ejh.13839] [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: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES This review discusses the role of immune dysfunction at the different stages of MM. METHODS Narrative review RESULTS: Multiple myeloma (MM) is a complex disease and immune dysfunction has been known to play an important role in disease pathogenesis, progression, and drug resistance. MM is known to be preceded by asymptomatic precursor states and progression from the precursor states to MM is likely related to a progressive impairment of the immune system. CONCLUSIONS An understanding of the role of the immune system in the progression of MM is important to guide the development of immunotherapeutic strategies for this disease.
Collapse
Affiliation(s)
- Cinnie Yentia Soekojo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System
| | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System
| |
Collapse
|
75
|
Invariant NKT cells dictate antitumor immunity elicited by a bispecific antibody cotargeting CD3 and BCMA. Blood Adv 2022; 6:5165-5170. [PMID: 35830292 DOI: 10.1182/bloodadvances.2022008118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/28/2022] [Indexed: 11/20/2022] Open
Abstract
CD3-engaging bispecific antibodies (BsAbs) have emerged as powerful therapeutic approaches by their ability to redirect T cells to eliminate tumor cells in an MHC-independent manner. However, it remains largely unknown how we can potentiate the efficacy of BsAbs. To address this, we investigated immunological mechanisms of action of a BsAb co-targeting CD3 and B-cell maturation antigen (BCMA) in syngeneic preclinical myeloma models. Treatment with the CD3/BCMA BsAb stimulated multiple CD3-expressing T cell subsets, as well as natural killer (NK) cells in the myeloma bone marrow (BM), highlighting its broad immunostimulatory effect. Notably, the BsAb-mediated immunostimulatory and anti-tumor effects were abrogated in mice lacking invariant NKT (iNKT) cells. Mechanistically, activation of iNKT cells and IL-12 production from dendritic cells (DCs) were crucial upstream events for triggering effective anti-tumor immunity by the BsAb. Myeloma progression was associated with reduced numbers of BM iNKT cells. Importantly, the therapeutic efficacy of a single dose of the CD3/BCMA BsAb was remarkably augmented by restoring iNKT cell activity using adoptive transfer of α-galactosylceramide-loaded DCs. Together, these results reveal iNKT cells as a critical player for the anti-tumor activity of CD3-engaging BsAbs, providing important translational implications.
Collapse
|
76
|
Davies FE, Pawlyn C, Usmani SZ, San-Miguel JF, Einsele H, Boyle EM, Corre J, Auclair D, Cho HJ, Lonial S, Sonneveld P, Stewart AK, Bergsagel PL, Kaiser MF, Weisel K, Keats JJ, Mikhael JR, Morgan KE, Ghobrial IM, Orlowski RZ, Landgren CO, Gay F, Caers J, Chng WJ, Chari A, Walker BA, Kumar SK, Costa LJ, Anderson KC, Morgan GJ. Perspectives on the Risk-Stratified Treatment of Multiple Myeloma. Blood Cancer Discov 2022; 3:273-284. [PMID: 35653112 PMCID: PMC9894570 DOI: 10.1158/2643-3230.bcd-21-0205] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The multiple myeloma treatment landscape has changed dramatically. This change, paralleled by an increase in scientific knowledge, has resulted in significant improvement in survival. However, heterogeneity remains in clinical outcomes, with a proportion of patients not benefiting from current approaches and continuing to have a poor prognosis. A significant proportion of the variability in outcome can be predicted on the basis of clinical and biochemical parameters and tumor-acquired genetic variants, allowing for risk stratification and a more personalized approach to therapy. This article discusses the principles that can enable the rational and effective development of therapeutic approaches for high-risk multiple myeloma.
Collapse
Affiliation(s)
| | - Charlotte Pawlyn
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital, Department of Haematology, London, United Kingdom
| | - Saad Z. Usmani
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Jill Corre
- Unité de Génomique du Myélome, Institut Universitaire du Cancer, Toulouse France. Institut National de la Santé et de la Recherche Médicale, Paris, France
| | - Daniel Auclair
- The Multiple Myeloma Research Foundation, Norwalk, Connecticut
| | - Hearn Jay Cho
- The Multiple Myeloma Research Foundation, Norwalk, Connecticut
- Multiple Myeloma Center of Excellence, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Pieter Sonneveld
- Erasmus MC Cancer Institute, Department of Hematology, Rotterdam, the Netherlands
| | - A. Keith Stewart
- University Health Network and the Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | - Martin F. Kaiser
- The Royal Marsden Hospital, Department of Haematology, London, United Kingdom
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
| | - Katja Weisel
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonathan J. Keats
- Integrated Cancer Genomics, Translational Genomics Research Institute, Phoenix, Arizona
| | - Joseph R. Mikhael
- Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, Arizona
| | | | - Irene M. Ghobrial
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Robert Z. Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - C. Ola Landgren
- Myeloma Program, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Francesca Gay
- Division of Hematology, University of Torino, Torino, Italy
| | - Joseph Caers
- Department of Hematology, Centre Hospitalier Universitaire (CHU) de Liège, Liège, Belgium
| | - Wee Joo Chng
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University, Indianapolis, Indiana
- Department of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ajai Chari
- Multiple Myeloma Center of Excellence, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Brian A. Walker
- Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology Oncology, Indiana University, Indianapolis, Indiana
| | - Shaji K. Kumar
- Department of Hematology, Mayo Clinic, Rochester, Minnesota
| | - Luciano J. Costa
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kenneth C. Anderson
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
77
|
Forster S, Radpour R. Molecular Impact of the Tumor Microenvironment on Multiple Myeloma Dissemination and Extramedullary Disease. Front Oncol 2022; 12:941437. [PMID: 35847862 PMCID: PMC9284036 DOI: 10.3389/fonc.2022.941437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/06/2022] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the most common malignant monoclonal disease of plasma cells. Aside from classical chemotherapy and glucocorticoids, proteasome inhibitors, immunomodulatory agents and monoclonal antibodies are used in the current treatment scheme of MM. The tumor microenvironment (TME) plays a fundamental role in the development and progression of numerous solid and non-solid cancer entities. In MM, the survival and expansion of malignant plasma cell clones heavily depends on various direct and indirect signaling pathways provided by the surrounding bone marrow (BM) niche. In a number of MM patients, single plasma cell clones lose their BM dependency and are capable to engraft at distant body sites or organs. The resulting condition is defined as an extramedullary myeloma (EMM). EMMs are highly aggressive disease stages linked to a dismal prognosis. Emerging literature demonstrates that the dynamic interactions between the TME and malignant plasma cells affect myeloma dissemination. In this review, we aim to summarize how the cellular and non-cellular BM compartments can promote plasma cells to exit their BM niche and metastasize to distant intra-or extramedullary locations. In addition, we list selected therapy concepts that directly target the TME with the potential to prevent myeloma spread.
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
- *Correspondence: Ramin Radpour,
| |
Collapse
|
78
|
From single-omics to interactomics: How can ligand-induced perturbations modulate single-cell phenotypes? ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 131:45-83. [PMID: 35871896 DOI: 10.1016/bs.apcsb.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cells suffer from perturbations by different stimuli, which, consequently, rise to individual alterations in their profile and function that may end up affecting the tissue as a whole. This is no different if we consider the effect of a therapeutic agent on a biological system. As cells are exposed to external ligands their profile can change at different single-omics levels. Detecting how these changes take place through different sequencing technologies is key to a better understanding of the effects of therapeutic agents. Single-cell RNA-sequencing stands out as one of the most common approaches for cell profiling and perturbation analysis. As a result, single-cell transcriptomics data can be integrated with other omics data sources, such as proteomics and epigenomics data, to clarify the perturbation effects and mechanism at the cell level. Appropriate computational tools are key to process and integrate the available information. This chapter focuses on the recent advances on ligand-induced perturbation and single-cell omics computational tools and algorithms, their current limitations, and how the deluge of data can be used to improve the current process of drug research and development.
Collapse
|
79
|
Pancancer Analyses Reveal Genomics and Clinical Characteristics of the SETDB1 in Human Tumors. JOURNAL OF ONCOLOGY 2022; 2022:6115878. [PMID: 35656340 PMCID: PMC9152430 DOI: 10.1155/2022/6115878] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/17/2022] [Indexed: 12/03/2022]
Abstract
Background. Malignant tumor is one of the most common diseases that seriously affect human health. The prior literature has reported the biological function and potential therapeutic targets of SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) as an oncogene. However, SETDB1 has rarely been analyzed from a pan-cancer perspective. Methods. Bioinformatics analysis tools and databases, including GeneCards, National Center for Biotechnology Information (NCBI), UniProt, Illustrator for Biological Sequences (IBS), Human Protein Atlas (HPA), GEPIA, TIMER2, Sangerbox 3.0, UALCAN, Kaplan-Meier (K-M) plotter, cBioPortal, Catalogue Of Somatic Mutations In Cancer (COSMIC), PhosphoSitePlus, TISIDB, STRING, and GeneMANIA, were utilized to clarify the biological functions and clinical significance of SETDB1 from a pan-cancer perspective. Results. In this study, the pan-cancer analysis demonstrated that SETDB1 showed significantly differential expression in most tumor tissues and paracancerous tissues, and SETDB1 expression was associated with clinicopathological features and clinical prognosis. We also found that SETDB1 mutations occurred in most tumors and were related to tumorigenesis. In addition, DNA methylation of SETDB1 primarily occurred at the cg10444928 site and was associated with prognosis in several human tumors. The predicted phosphorylation site of SETDB1 was Ser1006. We found that SETDB1 was significantly related to the specific tumor-infiltrating immune cell populations and expression of clinically targetable immune checkpoints and may be a promising immunotherapy target. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses also indicated that SETDB1 may function as crucial regulator in carcinogenesis of human cancers. Conclusions. SETDB1 is an important oncogene involved in tumorigenesis and tumor progression through different biological mechanisms. Furthermore, SETDB1 may be a potential therapeutic target for cancer treatment.
Collapse
|
80
|
He H, Li Z, Lu J, Qiang W, Jiang S, Xu Y, Fu W, Zhai X, Zhou L, Qian M, Du J. Single-cell RNA-seq reveals clonal diversity and prognostic genes of relapsed multiple myeloma. Clin Transl Med 2022; 12:e757. [PMID: 35297204 PMCID: PMC8926895 DOI: 10.1002/ctm2.757] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/15/2022] [Accepted: 02/21/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a clinically and biologically heterogeneous plasma-cell malignancy. Despite extensive research, disease heterogeneity and relapse remain a big challenge in MM therapeutics. We tried to dissect this disease and identify novel biomarkers for patient stratification and treatment outcome prediction by applying single-cell technology. METHODS We performed single-cell RNA sequencing (scRNA-seq) and variable-diversity-joining regions-targeted sequencing (scVDJ-seq) concurrently on bone marrow samples from a cohort of 18 patients with newly diagnosed MM (NDMM; n = 12) or refractory/relapsed MM (RRMM; n = 6). We analysed the malignant clonotypes using scVDJ-seq data and conducted data integration and cell-type annotation through the CCA algorithm based on gene expression profiling. Furthermore, we identified disease status-specific genes and modules by comparison of NDMM and RRMM datasets and explored the findings in a larger MM cohort from the MMRF CoMMpass study. RESULTS We found that all the myeloma cells in either diagnosed or relapsed samples were dominated by a major clone, with a few subclones in several samples (n = 5). Next, we investigated the universal transcriptional features of myeloma cells and identified eight meta-programs correlated with this disease, especially meta-programs 1 and 8 (M1 and M8), which were the most significant and related to cell cycle and stress response, respectively. Furthermore, we classified the malignant plasma cells into eight clusters and found that the cell numbers in clusters 2/6/7 were exclusively higher in relapsed samples. Besides, we identified several attractive candidates for biomarkers (e.g. SMAD1 and STMN1) associated with disease progression and relapse in our dataset and related to overall survival in the CoMMpass dataset. CONCLUSIONS Our data provide insights into the heterogeneity of MM as well as highlight the relevance of intra-tumour heterogeneity and discover novel biomarkers that might be a potent therapy.
Collapse
Affiliation(s)
- Haiyan He
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Zifeng Li
- Institute of Pediatrics and Department of Hematology and OncologyChildren's Hospital of Fudan UniversityNational Children's Medical CenterShanghaiChina
| | - Jing Lu
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Wanting Qiang
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Sihan Jiang
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Yaochen Xu
- Shanghai Key Laboratory of Medical Epigenetics, International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Weijun Fu
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Xiaowen Zhai
- Institute of Pediatrics and Department of Hematology and OncologyChildren's Hospital of Fudan UniversityNational Children's Medical CenterShanghaiChina
| | - Lin Zhou
- Department of Laboratory MedicineChangzheng HospitalNaval Medical UniversityShanghaiChina
| | - Maoxiang Qian
- Institute of Pediatrics and Department of Hematology and OncologyChildren's Hospital of Fudan UniversityNational Children's Medical CenterShanghaiChina
| | - Juan Du
- Department of HematologyMyeloma & Lymphoma CenterChangzheng HospitalNaval Medical UniversityShanghaiChina
| |
Collapse
|