1
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
2
|
Tan Y, Ren M, Hou J, Hou T, Lin X. Increased CD14 +HLA-DR -/low myeloid-derived suppressor cells can be regarded as a biomarker on disease severity and response to therapy in acute coronary syndrome. PeerJ 2024; 12:e18154. [PMID: 39399429 PMCID: PMC11468897 DOI: 10.7717/peerj.18154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/01/2024] [Indexed: 10/15/2024] Open
Abstract
Purpose This study aimed to investigate the dynamic changes in monocytic myeloid-derived suppressor cells (M-MDSCs) and their implications in the pathogenesis of acute coronary syndrome (ACS), shedding light on potential therapeutic targets. Experimental Design Peripheral blood samples were collected from 68 ACS patients, 35 stable angina pectoris (SAP) patients, and 30 healthy controls (HC). Multi-parameter flow cytometry was employed for analysis of M-MDSCs, explored with disease characteristics and progression. Results ACS patients exhibited an increased frequency of circulating M-MDSCs compared to SAP patients and HC. M-MDSCs levels demonstrated associations with ACS type, coronary artery lesions, multi-vessel disease, and cardiac dysfunction severity. Higher M-MDSCs levels were found in obese patients. Notably, therapy led to a significant decrease in M-MDSCs frequency. Furthermore, ACS patients exhibited elevated levels of interleukin (IL)-6, IL-10, granulocyte-macrophage colony-stimulating factor (GM-CSF), and tumor necrosis factor-α (TNF-α) in the cytokine profile associated with M-MDSCs. Increased expression of arginase-1(Arg-1) was observed in ACS patients, with positive correlations between M-MDSCs levels and IL-6, GM-CSF, and Arg-1 expression. The diagnostic performance of triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), and M-MDSCs levels varied in predicting the severity of coronary artery stenosis, with TG showing higher specificity, HDL-C displaying higher sensitivity, and M-MDSCs levels demonstrating balanced sensitivity and specificity. Conclusions Assessment of M-MDSCs frequency holds promise as a predictive marker for disease progression and therapy response of coronary artery stenosis. The elevated presence of M-MDSCs suggests their potential role in modulating ACS-related inflammation.
Collapse
Affiliation(s)
- Yinsheng Tan
- Department of Cardiovascular, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
- Department of Cardiovascular, East District of First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Mingde Ren
- Department of Cardiovascular, Lu’an People’s Hospital of Anhui Province, Lu’an, Anhui Province, China
| | - Jie Hou
- Department of Cardiovascular, East District of First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Tao Hou
- Department of Cardiovascular, East District of First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xianhe Lin
- Department of Cardiovascular, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
3
|
Arunachalam AK, Grégoire C, Coutinho de Oliveira B, Melenhorst JJ. Advancing CAR T-cell therapies: Preclinical insights and clinical translation for hematological malignancies. Blood Rev 2024:101241. [PMID: 39289094 DOI: 10.1016/j.blre.2024.101241] [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: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved significant success in achieving durable and potentially curative responses in patients with hematological malignancies. CARs are tailored fusion proteins that direct T cells to a specific antigen on tumor cells thereby eliciting a targeted immune response. The approval of several CD19-targeted CAR T-cell therapies has resulted in a notable surge in clinical trials involving CAR T cell therapies for hematological malignancies. Despite advancements in understanding response mechanisms, resistance patterns, and adverse events associated with CAR T-cell therapy, the translation of these insights into robust clinical efficacy has shown modest outcomes in both clinical trials and real-world scenarios. Therefore, the assessment of CAR T-cell functionality through rigorous preclinical studies plays a pivotal role in refining therapeutic strategies for clinical applications. This review provides an overview of the various in vitro and animal models used to assess the functionality of CAR T-cells. We discuss the findings from preclinical research involving approved CAR T-cell products, along with the implications derived from recent preclinical studies aiming to optimize the functionality of CAR T-cells. The review underscores the importance of robust preclinical evaluations and the need for models that accurately replicate human disease to bridge the gap between preclinical success and clinical efficacy.
Collapse
Affiliation(s)
- Arun K Arunachalam
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Céline Grégoire
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Beatriz Coutinho de Oliveira
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Jan Joseph Melenhorst
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
4
|
Aliazis K, Yenyuwadee S, Phikulsod P, Boussiotis VA. Emergency myelopoiesis in solid cancers. Br J Haematol 2024; 205:798-811. [PMID: 39044285 DOI: 10.1111/bjh.19656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
Cells of the innate and adaptive immune systems are the progeny of haematopoietic stem and progenitor cells (HSPCs). During steady-state myelopoiesis, HSPC undergo differentiation and proliferation but are called to respond directly and acutely to various signals that lead to emergency myelopoiesis, including bone marrow ablation, infections, and sterile inflammation. There is extensive evidence that many solid tumours have the potential to secrete classical myelopoiesis-promoting growth factors and other products able to mimic emergency haematopoiesis, and to aberrantly re-direct myeloid cell development into immunosuppressive cells with tumour promoting properties. Here, we summarize the current literature regarding the effects of solid cancers on HSPCs function and discuss how these effects might shape antitumour responses via a mechanism initiated at a site distal from the tumour microenvironment.
Collapse
Affiliation(s)
- Konstantinos Aliazis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sasitorn Yenyuwadee
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ployploen Phikulsod
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vassiliki A Boussiotis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Ask EH, Tschan-Plessl A, Hoel HJ, Kolstad A, Holte H, Malmberg KJ. MetaGate: Interactive analysis of high-dimensional cytometry data with metadata integration. PATTERNS (NEW YORK, N.Y.) 2024; 5:100989. [PMID: 39081571 PMCID: PMC11284499 DOI: 10.1016/j.patter.2024.100989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/12/2024] [Accepted: 04/15/2024] [Indexed: 08/02/2024]
Abstract
Flow cytometry is a powerful technology for high-throughput protein quantification at the single-cell level. Technical advances have substantially increased data complexity, but novel bioinformatical tools often show limitations in statistical testing, data sharing, cross-experiment comparability, or clinical data integration. We developed MetaGate as a platform for interactive statistical analysis and visualization of manually gated high-dimensional cytometry data with integration of metadata. MetaGate provides a data reduction algorithm based on a combinatorial gating system that produces a small, portable, and standardized data file. This is subsequently used to produce figures and statistical analyses through a fast web-based user interface. We demonstrate the utility of MetaGate through a comprehensive mass cytometry analysis of peripheral blood immune cells from 28 patients with diffuse large B cell lymphoma along with 17 healthy controls. Through MetaGate analysis, our study identifies key immune cell population changes associated with disease progression.
Collapse
Affiliation(s)
- Eivind Heggernes Ask
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- The Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Astrid Tschan-Plessl
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Hanna Julie Hoel
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Arne Kolstad
- Department of Oncology, Innlandet Hospital Trust Division Gjøvik, Lillehammer, Norway
| | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- The Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Sinkarevs S, Strumfs B, Volkova S, Strumfa I. Tumour Microenvironment: The General Principles of Pathogenesis and Implications in Diffuse Large B Cell Lymphoma. Cells 2024; 13:1057. [PMID: 38920685 PMCID: PMC11201569 DOI: 10.3390/cells13121057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is the most common type of non-Hodgkin lymphoma worldwide, constituting around 30-40% of all cases. Almost 60% of patients develop relapse of refractory DLBCL. Among the reasons for the therapy failure, tumour microenvironment (TME) components could be involved, including tumour-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), tumour-associated neutrophils (TANs), cancer-associated fibroblasts (CAFs), and different subtypes of cytotoxic CD8+ cells and T regulatory cells, which show complex interactions with tumour cells. Understanding of the TME can provide new therapeutic options for patients with DLBCL and improve their prognosis and overall survival. This review provides essentials of the latest understanding of tumour microenvironment elements and discusses their role in tumour progression and immune suppression mechanisms which result in poor prognosis for patients with DLBCL. In addition, we point out important markers for the diagnostic purposes and highlight novel therapeutic targets.
Collapse
Affiliation(s)
| | | | | | - Ilze Strumfa
- Department of Pathology, Riga Stradins University, 16 Dzirciema Street, LV-1007 Riga, Latvia
| |
Collapse
|
7
|
Pu LF, Li MM, Feng XJ, Zhang T, Hu LH, Zheng HM, Charwudzi A, Ding YY, Liu J, Liu ZL, Xiong SD. Accumulation of circulating myeloid-derived suppressor cell subsets: predicting poor clinical efficacy and prognosis through T cell suppression in non-Hodgkin's lymphoma. J Leukoc Biol 2024; 115:1094-1107. [PMID: 38369808 DOI: 10.1093/jleuko/qiae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/21/2023] [Accepted: 01/24/2024] [Indexed: 02/20/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are implicated in the regulation of immune responses closely associated with poor clinical outcomes in cancer. However, the MDSC subtypes in non-Hodgkin's lymphoma (NHL) have not been systematically investigated. So, we investigated the percentage of MDSC subsets in 78 newly diagnosed NHL patients by flow cytometry. The results showed that all MDSC subsets increased in NHL patients compared with healthy donors. Notably, MDSCs, monocytic MDSCs, and CD14 + CD66b + MDSCs significantly increased in NHL patients compared with those with lymphadenitis donors. polymorphonuclear MDSCs (PMN-MDSCs), early-stage MDSCs (e-MDSCs), and the International Prognostic Index were independent risk factors for poor clinical efficacy and were involved in constructing the nomogram for predicting clinical efficacy. Progression-free survival (PFS) was significantly shorter in patients with high level of MDSC subsets, and PMN-MDSCs emerged as an independent prognostic factor for PFS. PMN-MDSCs, e-MDSCs, and the International Prognostic Index were involved in constructing the nomogram for predicting PFS. Patients with a higher percentage of MDSCs, PMN-MDSCs, e-MDSCs, and CD14 + CD66b + MDSCs experienced a shorter overall survival compared with those with lower percentages. In addition, research on mechanisms found that T cell function was suppressed and mediated by the expansion of MDSCs via involving arginase-1 and interleukin-10 in vitro and in vivo. In conclusion, our study demonstrates that the increased circulating MDSC subsets predict poor clinical efficacy and prognosis in NHL, potentially involving T cell suppression through MDSC subset expansion. These findings indicate the potential of MDSC subsets as comprehensive diagnostic, prognostic biomarkers, and therapeutic targets for NHL.
Collapse
Affiliation(s)
- Lian-Fang Pu
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Man-Man Li
- Department of Hematology and Oncology, Minhang Hospital, Fudan University, Shanghai 201199, People's Republic of China
| | - Xiang-Jiang Feng
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Tun Zhang
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Lin-Hui Hu
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Hui-Min Zheng
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Alice Charwudzi
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Yang-Yang Ding
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Jun Liu
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Ze-Lin Liu
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| | - Shu-Dao Xiong
- Hematological Lab, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
- Research Center for Translational Medicine, The Second Hospital of Anhui Medical University, Hefei 230601, Anhui, People's Republic of China
| |
Collapse
|
8
|
Li MY, Chong LC, Duns G, Lytle A, Woolcock B, Jiang A, Telenius A, Ben-Neriah S, Nawaz W, Slack GW, Elisia I, Viganò E, Aoki T, Healy S, Krystal G, Venturutti L, Scott DW, Steidl C. TRAF3 loss-of-function reveals the noncanonical NF-κB pathway as a therapeutic target in diffuse large B cell lymphoma. Proc Natl Acad Sci U S A 2024; 121:e2320421121. [PMID: 38662551 PMCID: PMC11067025 DOI: 10.1073/pnas.2320421121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/29/2024] [Indexed: 05/05/2024] Open
Abstract
Here, we report recurrent focal deletions of the chr14q32.31-32 locus, including TRAF3, a negative regulator of NF-κB signaling, in de novo diffuse large B cell lymphoma (DLBCL) (24/324 cases). Integrative analysis revealed an association between TRAF3 copy number loss with accumulation of NIK, the central noncanonical (NC) NF-κB kinase, and increased NC NF-κB pathway activity. Accordingly, TRAF3 genetic ablation in isogenic DLBCL model systems caused upregulation of NIK and enhanced NC NF-κB downstream signaling. Knockdown or pharmacological inhibition of NIK in TRAF3-deficient cells differentially impaired their proliferation and survival, suggesting an acquired onco-addiction to NC NF-κB. TRAF3 ablation also led to exacerbated secretion of the immunosuppressive cytokine IL-10. Coculturing of TRAF3-deficient DLBCL cells with CD8+ T cells impaired the induction of Granzyme B and interferon (IFN) γ, which were restored following neutralization of IL-10. Our findings corroborate a direct relationship between TRAF3 genetic alterations and NC NF-κB activation, and highlight NIK as a potential therapeutic target in a defined subset of DLBCL.
Collapse
Affiliation(s)
- Michael Y. Li
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Lauren C. Chong
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Gerben Duns
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Andrew Lytle
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Bruce Woolcock
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Aixiang Jiang
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Adèle Telenius
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Susana Ben-Neriah
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Waqas Nawaz
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Graham W. Slack
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Ingrid Elisia
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Elena Viganò
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Tomohiro Aoki
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Shannon Healy
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Gerald Krystal
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - Leandro Venturutti
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
| | - David W. Scott
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| | - Christian Steidl
- Centre for Lymphoid Cancer, British Columbia Cancer, Vancouver, BCV5Z 1L3, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BCV6T 2B5, Canada
| |
Collapse
|
9
|
Zhang W, Liu M, Li W, Song Y. Immune cells in the B-cell lymphoma microenvironment: From basic research to clinical applications. Chin Med J (Engl) 2024; 137:776-790. [PMID: 38269619 PMCID: PMC10997228 DOI: 10.1097/cm9.0000000000002919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Indexed: 01/26/2024] Open
Abstract
ABSTRACT B-cell lymphoma is a group of hematological malignancies characterized by variable genetic and biological features and clinical behaviors. The tumor microenvironment (TME) is a complex network in tumors, which consists of surrounding blood vessels, extracellular matrix, immune and non-immune cells, and signaling molecules. Increasing evidence has shown that the TME, especially immune cells within, is a double-edged sword, acting either as a tumor killer or as a promoter of tumor progression. These pro-tumor activities are driven by subpopulations of immune cells that express typical markers but have unique transcriptional characteristics, making tumor-associated immune cells good targets for human anti-cancer therapy by ablating immunosuppressive cells or enhancing immune-activated cells. Thus, exploring the role of immune cells in the TME provides distinct insights for immunotherapy in B-cell lymphoma. In this review, we elucidated the interaction between immune cells and tumor cells and their function in the initiation, progression, and prognosis of B-cell lymphoma, from preclinical experiments to clinical trials. Furthermore, we outlined potential therapeutic approaches and discussed the potential clinical value and future perspectives of targeting immune cells in patients with B-cell lymphoma.
Collapse
Affiliation(s)
- Wenli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, Henan 450000, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mengmeng Liu
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, Henan 450000, China
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, Henan 450000, China
| |
Collapse
|
10
|
Zhuang Y, Chen J, Mai Z, Huang W, Zhong W. Signature Construction and Disulfidptosis-Related Molecular Cluster Identification for Better Prediction of Prognosis in Glioma. J Mol Neurosci 2024; 74:38. [PMID: 38573391 DOI: 10.1007/s12031-024-02216-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/24/2024] [Indexed: 04/05/2024]
Abstract
Disulfidptosis is a newly discovered form of regulatory cell death. However, the identification of disulfidptosis-related molecular subtypes and potential biomarkers in gliomas and their prognostic predictive potential need to be further elucidated. RNA sequencing profiles and the relevant clinical data were obtained from the Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). Disulfidptosis-related clusters were identified by unsupervised clustering analysis. Immune cell infiltration analysis and drug sensitivity analysis were used to explore the differences between clusters. Gene set enrichment analysis (GSEA) of differential genes between clusters was performed to explore the potential biological functions and signaling. A disulfidptosis-related scoring system (DRSS) was constructed based on a combined COX and LASSO analysis. Mendelian randomization (MR) analyses were used to further explore the causal relationship between levels of genes in DRSS and an increased risk of glioma. A prognosis nomogram was constructed based on the DRSS and 3 clinical features (age, WHO stage, and IDH status). The accuracy and stability of the prognosis nomogram were also validated in different cohorts. We identified two clusters that exhibited different prognoses, drug sensitivity profiles, and tumor microenvironment infiltration profiles. The overall survival (OS) of Cluster2 was significantly better than Cluster1. Cluster1 had an overall greater infiltration of immune cells compared to Cluster2. However, the Monocytes, activated B cells had higher infiltration abundance in Cluster2. GSEA results showed significant enrichment of immune-related biological processes in Cluster1, while Cluster2 was more enriched for functions related to neurotransmission and regulation. PER3, RAB34, NKX3-2, GPX7, FRA10AC1, and TGIF1 were finally included to construct DRSS. DRSS was independently related to prognosis. There was a significant difference in overall survival between the low-risk score group and the high-risk score group. Among six genes in DRSS, GPX7 levels were demonstrated to have a causal relationship with an increased risk of glioma. GPX7 may become a more promising biomarker for gliomas. The prognosis nomogram constructed based on the DRSS and three clinical features has considerable potential for predicting the prognosis of patients with glioma. Free online software for implementing this nomogram was established: https://yekun-zhuang.shinyapps.io/DynNomapp/ . Our study established a novel glioma classification based on the disulfidptosis-related molecular subtypes. We constructed the DRSS and the prognosis nomogram to accurately stratify the prognosis of glioma patients. GPX7 was identified as a more promising biomarker for glioma. We provide important insights into the treatment and prognosis of gliomas.
Collapse
Affiliation(s)
- Yekun Zhuang
- The Sixth Clinical Medical School, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China.
| | - Jiewen Chen
- The Sixth Clinical Medical School, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Zhuohao Mai
- The Sixth Clinical Medical School, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Wanting Huang
- The Sixth Clinical Medical School, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Wenyu Zhong
- The Sixth Clinical Medical School, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| |
Collapse
|
11
|
Tyrinova T, Batorov E, Aristova T, Ushakova G, Sizikova S, Denisova V, Chernykh E. Decreased circulating myeloid-derived suppressor cell count at the engraftment is one of the risk factors for multiple myeloma relapse after autologous hematopoietic stem cell transplantation. Heliyon 2024; 10:e26362. [PMID: 38434301 PMCID: PMC10907647 DOI: 10.1016/j.heliyon.2024.e26362] [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: 12/15/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024] Open
Abstract
Recent studies demonstrated that myeloid-derived suppressor cells (MDSCs) are involved in the pathogenesis and progression of multiple myeloma (MM). Nevertheless, data on the quantitative and functional changes in MDSCs during standard MM treatment remain poorly understood. Here, we determined that monocytic MDSCs (M-MDSC; CD14+HLA-DRlow/-) and granulocytic MDSCs (PMN-MDSC; Lin-HLA-DR-CD33+CD66b+) in MM patients in remission following induction therapy (IT) were significantly increased, while early MDSCs (E-MDSCs; Lin-HLA-DR-CD33+CD66b-) were decreased compared to the donor group. In progression, MM patients had the most pronounced decrease in E-MDSCs and enhanced levels of PMN-MDSCs. IT was accompanied with a decrease in the expression of arginase-1 (Arg-1). In MM patients with relapse or resistance to IT, Arg-1+ cell frequency in M-MDSCs and E-MDSCs, as well as PD-L1+ M-MDSCs, was increased, which may facilitate tumor immunosuppression. G-CSF administration led to a significant increment in the MDSC subsets. At the engraftment, circulating M-MDSC and PMN-MDSCs were temporarily increased, with a gradual decline to the pre-transplant levels in 12 months. The percentage of E-MDSCs was decreased at the leukocyte recovery. Patients with a higher (>Me) M-MDSC count at the engraftment had a shorter post-transplant leukopenia duration (Me 11 vs. 13 days; pU = 0.0086). The advanced MM stage, depth of response, and lower relative count of circulating E-MDSCs at the engraftment were independent risk factors associated with a lower progression-free survival. The obtained data allow us to hypothesize that MDSCs may play a positive role at the stage of leukocyte recovery by ameliorating the long-term anti-tumor response in MM.
Collapse
Affiliation(s)
- Tamara Tyrinova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| | - Egor Batorov
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| | - Tatyana Aristova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| | - Galina Ushakova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| | - Svetlana Sizikova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| | - Vera Denisova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| | - Elena Chernykh
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintscevskaya str., Novosibirsk, 630099, Russian Federation
| |
Collapse
|
12
|
Lin Q, Su J, Fang Y, Zhong Z, Chen J, Zhang C. S100A8 is a prognostic signature and associated with immune response in diffuse large B-cell lymphoma. Front Oncol 2024; 14:1344669. [PMID: 38361783 PMCID: PMC10867108 DOI: 10.3389/fonc.2024.1344669] [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: 11/26/2023] [Accepted: 01/12/2024] [Indexed: 02/17/2024] Open
Abstract
Background S100A8, a calcium-binding protein belonging to the S100 family, is involved in immune responses and multiple tumor pathogens. Diffuse large B-cell lymphoma (DLBCL) is one of the most common types of B-cell lymphoma and remains incurable in 40% of patients. However, the role of S100A8 and its regulation of the immune response in DLBCL remain unclear. Methods The differential expression of S100A8 was identified via the GEO and TCGA databases. The prognostic role of S100A8 in DLBCL was calculated using the Kaplan-Meier curve. The function enrichment of differentially expressed genes (DEGs) was explored through GO, KEGG, GSEA, and PPI analysis. In our cohort, the expression of S100A8 was verified. Meanwhile, the biological function of S100A8 was applied after the inhibition of S100A8 in an in vitro experiment. The association between S100A8 and immune cell infiltration and treatment response in DLBCL was analyzed. Results S100A8 was significantly overexpressed and related to a poor prognosis in DLBCL patients. Function enrichment analysis revealed that DEGs were mainly enriched in the IL-17 signaling pathway. Our cohort also verified this point. In vitro experiments suggested that inhibition of S100A8 should promote cell apoptosis and suppress tumor growth. Single-cell RNA sequence analysis indicated that S100A8 might be associated with features of the tumor microenvironment (TME), and immune infiltration analyses discovered that S100A8 expression was involved in TME. In terms of drug screening, we predicted that many drugs were associated with preferable sensitivity. Conclusion Elevated S100A8 expression is associated with a poor prognosis and immune infiltration in DLBCL. Inhibition of S100A8 could promote cell apoptosis and suppress tumor growth. Meanwhile, S100A8 has the potential to be a promising immunotherapeutic target for patients with DLBCL.
Collapse
Affiliation(s)
- Qi Lin
- Department of Pharmacy, The Affiliated Hospital of Putian University, Putian, Fujian, China
- Pharmaceutical and Medical Technology College, Putian University, Putian, Fujian, China
| | - Jianlin Su
- Pharmaceutical and Medical Technology College, Putian University, Putian, Fujian, China
| | - Yuanyuan Fang
- Pharmaceutical and Medical Technology College, Putian University, Putian, Fujian, China
| | - Zhihao Zhong
- Pharmaceutical and Medical Technology College, Putian University, Putian, Fujian, China
| | - Jie Chen
- Pharmaceutical and Medical Technology College, Putian University, Putian, Fujian, China
| | - Chaofeng Zhang
- Department of Hematology and Rheumatology, the Affiliated Hospital of Putian University, Putian, Fujian, China
| |
Collapse
|
13
|
Ventin M, Cattaneo G, Maggs L, Arya S, Wang X, Ferrone CR. Implications of High Tumor Burden on Chimeric Antigen Receptor T-Cell Immunotherapy: A Review. JAMA Oncol 2024; 10:115-121. [PMID: 37943567 DOI: 10.1001/jamaoncol.2023.4504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Importance Chimeric antigen receptor (CAR) T-cell therapy has redefined the therapeutic landscape of several hematologic malignant tumors. Despite its clinical efficacy, many patients with cancer experience nonresponse to CAR T-cell treatment, disease relapse within months, or severe adverse events. Furthermore, CAR T-cell therapy has demonstrated minimal to no clinical efficacy in the treatment of solid tumors in clinical trials. Observations A complex interplay between high tumor burden and the systemic and local tumor microenvironment on clinical outcomes of CAR T-cell therapy is emerging from preclinical and clinical data. The hallmarks of advanced cancers-namely, inflammation and immune dysregulation-sustain cancer progression. They negatively affect the production, expansion, antitumor activity, and persistence of CAR T-cell products. Understanding of CAR T-cell therapy, mechanisms underlying its failure, and adverse events under conditions of high tumor burden is critical for realizing the full potential of this novel treatment approach. Conclusions and Relevance This review focuses on linking the efficacy and safety of CAR T-cell therapy with tumor burden. Its limitations relative to high tumor burden, systemic inflammation, and immune dysregulation are discussed. Emerging clinical approaches to overcome these obstacles and more effectively incorporate this therapeutic strategy into the treatment paradigm of patients with solid malignant tumors are also described.
Collapse
Affiliation(s)
- Marco Ventin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Giulia Cattaneo
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Luke Maggs
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Shahrzad Arya
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Cristina R Ferrone
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
14
|
Zhang C, Lin Q, Li C, Qiu Y, Chen J, Zhu X. Comprehensive analysis of the prognostic implication and immune infiltration of CISD2 in diffuse large B-cell lymphoma. Front Immunol 2023; 14:1277695. [PMID: 38155967 PMCID: PMC10754510 DOI: 10.3389/fimmu.2023.1277695] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/15/2023] [Indexed: 12/30/2023] Open
Abstract
Background Diffuse large B-cell lymphoma (DLBCL) is the most common B-cell lymphoma in adults. CDGSH iron sulfur domain 2 (CISD2) is an iron-sulfur protein and plays a critical role of cell proliferation. The aberrant expression of CISD2 is associated with the progression of multiple cancers. However, its role in DLBCL remains unclear. Methods The differential expression of CISD2 was identified via public databases, and quantitative real-time PCR (qRT-PCR) and western blot were used to identifed the expression of CISD2. We estimated the impact of CISD2 on clinical prognosis using the Kaplan-Meier plotter. Meanwhile, the drug sensitivity of CISD2 was assessed using CellMiner database. The 100 CISD2-related genes from STRING obtained and analyzed using the LASSO Cox regression. A CISD2 related signature for risk model (CISD2Risk) was established. The PPI network of CISD2Risk was performed, and functional enrichment was conducted through the DAVID database. The impacts of CISD2Risk on clinical features were analyzed. ESTIMATE, CIBERSORT, and MCP-counter algorithm were used to identify CISD2Risk associated with immune infiltration. Subsequently, Univariate and multivariate Cox regression analysis were applied, and a prognostic nomogram, accompanied by a calibration curve, was constructed to predict 1-, 3-, and 5-years survival probabilities. Results CISD2 was upregulated in DLBCL patients comparing with normal controls via public datasets, similarly, CISD2 was highly expressed in DLBCL cell lines. Overexpression of CISD2 was associated with poor prognosis in DLBCL patients based on the GSE31312, the GSE32918, and GSE93984 datasets (P<0.05). Nine drugs was considered as a potential therapeutic agents for CISD2. By using the LASSO cox regression, twenty seven genes were identified to construct CISD2Risk, and biological functions of these genes might be involved in apoptosis and P53 signaling pathway. The high CISD2Risk value had a worse prognosis and therapeutic effect (P<0.05). The higher stromal score, immune score, and ESTIMATE score were associated with lowe CISD2Risk value, CISD2Risk was negatively correlated with several immune infiltrating cells (macrophages M0 and M1, CD8 T cells, CD4 naïve T cells, NK cell, etc) that might be correlated with better prognosis. Additionally, The high CISD2Risk was identified as an independent prognostic factor for DLBCL patients using both univariate and multivariate Cox regression. The nomogram produced accurate predictions and the calibration curves were in good agreement. Conclusion Our study demonstrates that high expression of CISD2 in DLBCL patients is associated with poor prognosis. We have successfully constructed and validated a good prognostic prediction and efficacy monitoring for CISD2Risk that included 27 genes. Meanwhile, CISD2Risk may be a promising evaluator for immune infiltration and serve as a reference for clinical decision-making in DLBCL patients.
Collapse
Affiliation(s)
- ChaoFeng Zhang
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
- Department of Hematology and Rheumatology, The Affiliated Hospital of Putian University, Putian, China
- The School of Basic Medicine, Putian University, Putian, China
| | - Qi Lin
- Department of Pharmacy, The Affiliated Hospital of Putian University, Putian, China
| | - ChunTuan Li
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yang Qiu
- The School of Basic Medicine, Putian University, Putian, China
| | - JingYu Chen
- The School of Basic Medicine, Putian University, Putian, China
| | - XiongPeng Zhu
- Department of Haematology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
15
|
Meng Y, Ye F, Nie P, Zhao Q, An L, Wang W, Qu S, Shen Z, Cao Z, Zhang X, Jiao S, Wu D, Zhou Z, Wei L. Immunosuppressive CD10 +ALPL + neutrophils promote resistance to anti-PD-1 therapy in HCC by mediating irreversible exhaustion of T cells. J Hepatol 2023; 79:1435-1449. [PMID: 37689322 DOI: 10.1016/j.jhep.2023.08.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/20/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND & AIMS Remodeling the tumor microenvironment is a critical strategy for treating advanced hepatocellular carcinoma (HCC). Yet, how distinct cell populations in the microenvironment mediate tumor resistance to immunotherapies, such as anti-PD-1, remains poorly understood. METHODS We analyzed the transcriptomic profile, at a single-cell resolution, of tumor tissues from patients with HCC scheduled to receive anti-PD-1-based immunotherapy. Our comparative analysis and experimental validation using flow cytometry and histopathological analysis uncovered a discrete subpopulation of cells associated with resistance to anti-PD-1 treatment in patients and a rat model. A TurboID-based proximity labeling approach was deployed to gain mechanistic insights into the reprogramming of the HCC microenvironment. RESULTS We identified CD10+ALPL+ neutrophils as being associated with resistance to anti-PD-1 treatment. These neutrophils exhibited a strong immunosuppressive activity by inducing an apparent "irreversible" exhaustion of T cells in terms of cell number, frequency, and gene profile. Mechanistically, CD10+ALPL+ neutrophils were induced by tumor cells, i.e., tumor-secreted NAMPT reprogrammed CD10+ALPL+ neutrophils through NTRK1, maintaining them in an immature state and inhibiting their maturation and activation. CONCLUSIONS Collectively, our results reveal a fundamental mechanism by which CD10+ALPL+ neutrophils contribute to tumor immune escape from durable anti-PD-1 treatment. These data also provide further insights into novel immunotherapy targets and possible synergistic treatment regimens. IMPACT AND IMPLICATIONS Herein, we discovered that tumor cells reprogrammed CD10+ALPL+ neutrophils to induce the "irreversible" exhaustion of T cells and hence allow tumors to escape from the intended effects of anti-PD-1 treatment. Our data provided a new theoretical basis for the elucidation of special cell populations and revealed a molecular mechanism underpinning resistance to immunotherapy. Targeting these cells alongside existing immunotherapy could be looked at as a potentially more effective therapeutic approach.
Collapse
Affiliation(s)
- Yan Meng
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China; Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Fei Ye
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China; School of Medicine, Shanghai University, Shanghai 200444, China
| | - Pingping Nie
- Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiudong Zhao
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Liwei An
- Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shuping Qu
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Zhemin Shen
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Zhifa Cao
- Department of Stomatology, Shanghai Tenth People's Hospital, Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaobing Zhang
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Dong Wu
- Department of Hepatic Surgery, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, Zhongshan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Third Affiliated Hospital of Second Military Medical University, 225 Changhai Road, Shanghai, 200438, China.
| |
Collapse
|
16
|
Ask EH, Tschan-Plessl A, Hoel HJ, Kolstad A, Holte H, Malmberg KJ. MetaGate: Interactive Analysis of High-Dimensional Cytometry Data with Meta Data Integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.27.564454. [PMID: 37961421 PMCID: PMC10634916 DOI: 10.1101/2023.10.27.564454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Flow cytometry is a powerful technology for high-throughput protein quantification at the single-cell level, widely used in basic research and routine clinical diagnostics. Traditionally, data analysis is carried out using manual gating, in which cut-offs are defined manually for each marker. Recent technical advances, including the introduction of mass cytometry, have increased the number of proteins that can be simultaneously assessed in each cell. To tackle the resulting escalation in data complexity, numerous new analysis algorithms have been developed. However, many of these show limitations in terms of providing statistical testing, data sharing, cross-experiment comparability integration with clinical data. We developed MetaGate as a platform for interactive statistical analysis and visualization of manually gated high-dimensional cytometry data with integration of clinical meta data. MetaGate allows manual gating to take place in traditional cytometry analysis software, while providing a combinatorial gating system for simple and transparent definition of biologically relevant cell populations. We demonstrate the utility of MetaGate through a comprehensive analysis of peripheral blood immune cells from 28 patients with diffuse large B-cell lymphoma (DLBCL) along with 17 age- and sex-matched healthy controls using two mass cytometry panels made of a total of 55 phenotypic markers. In a two-step process, raw data from 143 FCS files is first condensed through a data reduction algorithm and combined with information from manual gates, user-defined cellular populations and clinical meta data. This results in one single small project file containing all relevant information to allow rapid statistical calculation and visualization of any desired comparison, including box plots, heatmaps and volcano plots. Our detailed characterization of the peripheral blood immune cell repertoire in patients with DLBCL corroborate previous reports showing expansion of monocytic myeloid-derived suppressor cells, as well as an inverse correlation between NK cell numbers and disease progression.
Collapse
Affiliation(s)
- Eivind Heggernes Ask
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- The Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Astrid Tschan-Plessl
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| | - Hanna Julie Hoel
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Arne Kolstad
- Department of Oncology, Innlandet Hospital Trust Division Gjøvik, Lillehammer, Norway
| | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for B cell malignancies, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karl-Johan Malmberg
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- The Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Jiang W, Hu K, Liu X, Gao J, Zhu L. Single-cell transcriptome analysis reveals the clinical implications of myeloid-derived suppressor cells in head and neck squamous cell carcinoma. Pathol Oncol Res 2023; 29:1611210. [PMID: 37475874 PMCID: PMC10354270 DOI: 10.3389/pore.2023.1611210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/28/2023] [Indexed: 07/22/2023]
Abstract
Head and neck squamous cell carcinoma (HNSC) is the most common malignant tumor that arises in the epithelium of the head and neck regions. Myeloid-derived suppressor cells (MDSCs) are one of the tumor-infiltrating immune cell populations, which play a powerful role in inhibiting anti-tumor immune response. Herein, we employed a single-cell RNA sequencing (scRNA-seq) dataset to dissect the heterogeneity of myeloid cells. We found that SPP1 + tumor-associated macrophages (TAMs) and MDSCs were the most abundant myeloid cells in the microenvironment. By cell cluster deconvolution from bulk RNA-seq datasets of larger patient groups, we observed that highly-infiltrated MDSC was a poor prognostic marker for patients' overall survival (OS) probabilities. To better apply the MDSC OS prediction values, we identified a set of six MDSC-related genes (ALDOA, CD52, FTH1, RTN4, SLC2A3, and TNFAIP6) as the prognostic signature. In both training and test cohorts, MDSC-related prognostic signature showed a promising value for predicting patients' prognosis outcomes. Further parsing the ligand-receptor pairs of intercellular communications by CellChat, we found that MDSCs could frequently interact with cytotoxic CD8 + T cells, SPP1 + TAMs, and endothelial cells. These interactions likely contributed to the establishment of an immunosuppressive microenvironment and the promotion of tumor angiogenesis. Our findings suggest that targeting MDSCs may serve as an alternative and promising target for the immunotherapy of HNSC.
Collapse
Affiliation(s)
- Wenru Jiang
- Department of Implant and Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kangyao Hu
- Department of Implant and Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaofei Liu
- Department of Implant and Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jili Gao
- Department of Implant and Prosthodontics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liping Zhu
- Department of Implant and Prosthodontics, Harbin First Hospital, Harbin, China
| |
Collapse
|
18
|
Cholujova D, Beke G, Hunter ZR, Hideshima T, Flores L, Zeleznikova T, Harrachova D, Klucar L, Leiba M, Drgona L, Treon SP, Kastritis E, Dorfman DM, Anderson KC, Jakubikova J. Dysfunctions of innate and adaptive immune tumor microenvironment in Waldenström macroglobulinemia. Int J Cancer 2023; 152:1947-1963. [PMID: 36533670 PMCID: PMC9992277 DOI: 10.1002/ijc.34405] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Waldenström macroglobulinemia (WM) is a rare subtype of non-Hodgkin lymphoma characterized by malignant lymphoplasmacytic cells in the bone marrow (BM). To dissect the pathophysiology of WM, we evaluated clonal cells by mapping of B cell lymphomagenesis with adaptive and innate immune tumor microenvironment (TME) in the BM of WM patients using mass cytometry (CyTOF). In-depth immunophenotypic profiling of WM cells exhibited profound expansion of clonal cells in both unswitched and switched memory B cells and also plasma cells with aberrant expression variations. WM B lymphomagenesis was associated with reduction of most B cell precursors assessed with the same clonally restricted light chain and phenotypic changes. The immune TME was infiltrated by mature monocytes, neutrophils and adaptive T cells, preferentially subsets of effector T helper, effector CTL and effector memory CTL cells that were associated with superior overall survival (OS), in contrast to progenitors of T cells and myeloid/monocytic lineage subsets that were suppressed in WM cohort. Moreover, decrease in immature B and NKT cells was related to worse OS in WM patients. Innate and adaptive immune subsets of WM TME were modulated by immune checkpoints, including PD-1/PD-L1&PD-L2, TIGIT/PVR, CD137/CD137-L, CTLA-4, BTLA and KIR expression. The response of ibrutinib treatment to the reduction of clonal memory B cell was associated with high levels of immature B cells and effector memory CTL cells. Our study demonstrates that CyTOF technology is a powerful approach for characterizing the pathophysiology of WM at various stages, predicting patient risk and monitoring the effectiveness of treatment strategies.
Collapse
Affiliation(s)
- Danka Cholujova
- Department of Tumor Immunology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Centre for Advanced Materials Application, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Gabor Beke
- Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Zachary R Hunter
- Bing Center for Waldenström Macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Teru Hideshima
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Boston, Massachusetts, USA
| | - Ludmila Flores
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Boston, Massachusetts, USA
| | - Tatiana Zeleznikova
- Department of Oncohematology, St. Elizabeth Cancer Institute Hospital, Bratislava, Slovakia
| | - Denisa Harrachova
- Department of Oncohematology, Hospital Cyril and Methodius, Bratislava, Slovakia
| | - Lubos Klucar
- Institute of Molecular Biology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Merav Leiba
- Assuta Ashdod University Hospital, Faculty of Health Science, Ben-Gurion University of the Negev, Negev, Israel
| | - Lubos Drgona
- Department of Oncohematology, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Steven P Treon
- Bing Center for Waldenström Macroglobulinemia, Dana Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kenneth C Anderson
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Boston, Massachusetts, USA
| | - Jana Jakubikova
- Department of Tumor Immunology, Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Centre for Advanced Materials Application, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Jerome Lipper Multiple Myeloma Center, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Bhardwaj V, Ansell SM. Modulation of T-cell function by myeloid-derived suppressor cells in hematological malignancies. Front Cell Dev Biol 2023; 11:1129343. [PMID: 37091970 PMCID: PMC10113446 DOI: 10.3389/fcell.2023.1129343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are pathologically activated neutrophils and monocytes that negatively regulate the immune response to cancer and chronic infections. Abnormal myelopoiesis and pathological activation of myeloid cells generate this heterogeneous population of myeloid-derived suppressor cells. They are characterized by their distinct transcription, phenotypic, biochemical, and functional features. In the tumor microenvironment (TME), myeloid-derived suppressor cells represent an important class of immunosuppressive cells that correlate with tumor burden, stage, and a poor prognosis. Myeloid-derived suppressor cells exert a strong immunosuppressive effect on T-cells (and a broad range of other immune cells), by blocking lymphocyte homing, increasing production of reactive oxygen and nitrogen species, promoting secretion of various cytokines, chemokines, and immune regulatory molecules, stimulation of other immunosuppressive cells, depletion of various metabolites, and upregulation of immune checkpoint molecules. Additionally, the heterogeneity of myeloid-derived suppressor cells in cancer makes their identification challenging. Overall, they serve as a major obstacle for many cancer immunotherapies and targeting them could be a favorable strategy to improve the effectiveness of immunotherapeutic interventions. However, in hematological malignancies, particularly B-cell malignancies, the clinical outcomes of targeting these myeloid-derived suppressor cells is a field that is still to be explored. This review summarizes the complex biology of myeloid-derived suppressor cells with an emphasis on the immunosuppressive pathways used by myeloid-derived suppressor cells to modulate T-cell function in hematological malignancies. In addition, we describe the challenges, therapeutic strategies, and clinical relevance of targeting myeloid-derived suppressor cells in these diseases.
Collapse
|
20
|
Wang S, Zhao X, Wu S, Cui D, Xu Z. Myeloid-derived suppressor cells: key immunosuppressive regulators and therapeutic targets in hematological malignancies. Biomark Res 2023; 11:34. [PMID: 36978204 PMCID: PMC10049909 DOI: 10.1186/s40364-023-00475-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
The immunosuppressive tumor microenvironment (TME) supports the development of tumors and limits tumor immunotherapy, including hematological malignancies. Hematological malignancies remain a major public health issue with high morbidity and mortality worldwide. As an important component of immunosuppressive regulators, the phenotypic characteristics and prognostic value of myeloid-derived suppressor cells (MDSCs) have received much attention. A variety of MDSC-targeting therapeutic approaches have produced encouraging outcomes. However, the use of various MDSC-targeted treatment strategies in hematologic malignancies is still difficult due to the heterogeneity of hematologic malignancies and the complexity of the immune system. In this review, we summarize the biological functions of MDSCs and further provide a summary of the phenotypes and suppressive mechanisms of MDSC populations expanded in various types of hematological malignancy contexts. Moreover, we discussed the clinical correlation between MDSCs and the diagnosis of malignant hematological disease, as well as the drugs targeting MDSCs, and focused on summarizing the therapeutic strategies in combination with other immunotherapies, such as various immune checkpoint inhibitors (ICIs), that are under active investigation. We highlight the new direction of targeting MDSCs to improve the therapeutic efficacy of tumors.
Collapse
Affiliation(s)
- Shifen Wang
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyun Zhao
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siwen Wu
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Zhenshu Xu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
21
|
Stavrou V, Fultang L, Booth S, De Simone D, Bartnik A, Scarpa U, Gneo L, Panetti S, Potluri S, Almowaled M, Barlow J, Jankevics A, Lloyd G, Southam A, Priestman DA, Cheng P, Dunn W, Platt F, Endou H, Craddock C, Keeshan K, Mussai F, De Santo C. Invariant NKT cells metabolically adapt to the acute myeloid leukaemia environment. Cancer Immunol Immunother 2023; 72:543-560. [PMID: 35962843 PMCID: PMC9947083 DOI: 10.1007/s00262-022-03268-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Acute myeloid leukaemia (AML) creates an immunosuppressive environment to conventional T cells through Arginase 2 (ARG2)-induced arginine depletion. We identify that AML blasts release the acute phase protein serum amyloid A (SAA), which acts in an autocrine manner to upregulate ARG2 expression and activity, and promote AML blast viability. Following in vitro cross-talk invariant natural killer T (iNKT) cells become activated, upregulate mitochondrial capacity, and release IFN-γ. iNKT retain their ability to proliferate and be activated despite the low arginine AML environment, due to the upregulation of Large Neutral Amino Acid Transporter-1 (LAT-1) and Argininosuccinate Synthetase 1 (ASS)-dependent amino acid pathways, resulting in AML cell death. T cell proliferation is restored in vitro and in vivo. The capacity of iNKT cells to restore antigen-specific T cell immunity was similarly demonstrated against myeloid-derived suppressor cells (MDSCs) in wild-type and Jα18-/- syngeneic lymphoma-bearing models in vivo. Thus, stimulation of iNKT cell activity has the potential as an immunotherapy against AML or as an adjunct to boost antigen-specific T cell immunotherapies in haematological or solid cancers.
Collapse
Affiliation(s)
- Victoria Stavrou
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Livingstone Fultang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Sarah Booth
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Daniele De Simone
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Arekdiusz Bartnik
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ugo Scarpa
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Luciana Gneo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Silvia Panetti
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Sandeep Potluri
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Meaad Almowaled
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, G12 0YN, UK
| | - Jonathan Barlow
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Andris Jankevics
- School of Biosciences and Phenome Centre Birmingham, University of Birmingham, Birmingham, B15 2TT, UK
| | - Gavin Lloyd
- School of Biosciences and Phenome Centre Birmingham, University of Birmingham, Birmingham, B15 2TT, UK
| | - Andrew Southam
- School of Biosciences and Phenome Centre Birmingham, University of Birmingham, Birmingham, B15 2TT, UK
| | - David A Priestman
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Paul Cheng
- Bio-Cancer Treatment International, Hong Kong Science Park, Hong Kong, China
| | - Warwick Dunn
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, B15 2TT, UK.,Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Frances Platt
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Hitoshi Endou
- J-Pharma Co. Ltd, Yokohama, Kanagawa, 230-0046, Japan
| | - Charles Craddock
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, B15 2TT, UK
| | - Karen Keeshan
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Glasgow, G12 0YN, UK
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
22
|
Ng WL, Ansell SM, Mondello P. Insights into the tumor microenvironment of B cell lymphoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:362. [PMID: 36578079 PMCID: PMC9798587 DOI: 10.1186/s13046-022-02579-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022]
Abstract
The standard therapies in lymphoma have predominantly focused on targeting tumor cells with less of a focus on the tumor microenvironment (TME), which plays a critical role in favoring tumor growth and survival. Such an approach may result in increasingly refractory disease with progressively reduced responses to subsequent treatments. To overcome this hurdle, targeting the TME has emerged as a new therapeutic strategy. The TME consists of T and B lymphocytes, tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and other components. Understanding the TME can lead to a comprehensive approach to managing lymphoma, resulting in therapeutic strategies that target not only cancer cells, but also the supportive environment and thereby ultimately improve survival of lymphoma patients. Here, we review the normal function of different components of the TME, the impact of their aberrant behavior in B cell lymphoma and the current TME-direct therapeutic avenues.
Collapse
Affiliation(s)
- Wern Lynn Ng
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Stephen M. Ansell
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Patrizia Mondello
- grid.66875.3a0000 0004 0459 167XDivision of Hematology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| |
Collapse
|
23
|
Tumino N, Fiore PF, Pelosi A, Moretta L, Vacca P. Myeloid derived suppressor cells in tumor microenvironment: Interaction with innate lymphoid cells. Semin Immunol 2022; 61-64:101668. [PMID: 36370673 DOI: 10.1016/j.smim.2022.101668] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/10/2022]
Abstract
Human myeloid-derived suppressor cells (MDSC) represent a stage of immature myeloid cells and two main subsets can be identified: monocytic and polymorphonuclear. MDSC contribute to the establishment of an immunosuppressive tumor microenvironment (TME). The presence and the activity of MDSC in patients with different tumors correlate with poor prognosis. As previously reported, MDSC promote tumor growth and use different mechanisms to suppress the immune cell-mediated anti-tumor activity. Immunosuppression mechanisms used by MDSC are broad and depend on their differentiation stage and on the pathological context. It is known that some effector cells of the immune system can play an important role in the control of tumor progression and metastatic spread. In particular, innate lymphoid cells (ILC) contribute to control tumor growth representing a potential, versatile and, immunotherapeutic tool. Despite promising results obtained by using new cellular immunotherapeutic approaches, a relevant proportion of patients do not benefit from these therapies. Novel strategies have been investigated to overcome the detrimental effect exerted by the immunosuppressive component of TME (i.e. MDSC). In this review, we summarized the characteristics and the interactions occurring between MDSC and ILC in different tumors discussing how a deeper knowledge on MDSC biology could represent an important target for tumor immunotherapy capable of decreasing immunosuppression and enhancing anti-tumor activity exerted by immune cells.
Collapse
Affiliation(s)
- Nicola Tumino
- Innate lymphoid cells Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, Rome, Italy.
| | | | - Andrea Pelosi
- Tumor Immunology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Lorenzo Moretta
- Tumor Immunology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Paola Vacca
- Innate lymphoid cells Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| |
Collapse
|
24
|
Fan R, De Beule N, Maes A, De Bruyne E, Menu E, Vanderkerken K, Maes K, Breckpot K, De Veirman K. The prognostic value and therapeutic targeting of myeloid-derived suppressor cells in hematological cancers. Front Immunol 2022; 13:1016059. [PMID: 36304465 PMCID: PMC9592826 DOI: 10.3389/fimmu.2022.1016059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022] Open
Abstract
The success of immunotherapeutic approaches in hematological cancers is partially hampered by the presence of an immunosuppressive microenvironment. Myeloid-derived suppressor cells (MDSC) are key components of this suppressive environment and are frequently associated with tumor cell survival and drug resistance. Based on their morphology and phenotype, MDSC are commonly subdivided into polymorphonuclear MDSC (PMN-MDSC or G-MDSC) and monocytic MDSC (M-MDSC), both characterized by their immunosuppressive function. The phenotype, function and prognostic value of MDSC in hematological cancers has been intensively studied; however, the therapeutic targeting of this cell population remains challenging and needs further investigation. In this review, we will summarize the prognostic value of MDSC and the different attempts to target MDSC (or subtypes of MDSC) in hematological cancers. We will discuss the benefits, challenges and opportunities of using MDSC-targeting approaches, aiming to enhance anti-tumor immune responses of currently used cellular and non-cellular immunotherapies.
Collapse
Affiliation(s)
- Rong Fan
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nathan De Beule
- Department of Clinical Hematology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Anke Maes
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Center for Medical Genetics, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- *Correspondence: Kim De Veirman,
| |
Collapse
|
25
|
Jurgec S, Jezernik G, Gorenjak M, Büdefeld T, Potočnik U. Meta-Analytic Comparison of Global RNA Transcriptomes of Acute and Chronic Myeloid Leukemia Cells Reveals Novel Gene Candidates Governing Myeloid Malignancies. Cancers (Basel) 2022; 14:cancers14194681. [PMID: 36230605 PMCID: PMC9562668 DOI: 10.3390/cancers14194681] [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: 09/02/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Despite advances in the understanding of genetic risk factors and molecular mechanisms underlying acute myeloid leukemia (AML) and chronic myeloid leukemia (CML), clinical outcomes of current therapies in terms of disease relapse and mortality rate pose a great economic and social burden. To overcome this, the identification of new molecular prognostic biomarkers and pharmacological targets is crucial. Recent studies have suggested that AML and CML may share common pathogenic mechanisms and cellular substrates. To this end, in the present study, global transcriptome profiles of AML and CML at the molecular and cellular level were directly compared using a combination of meta-analysis and modern statistics, and novel candidate genes and specific biological processes associated with the pathogenesis of AML and CML were characterized. Our study significantly improves our current understanding of myeloid leukemia and will help develop new therapeutic targets and biomarkers for disease progression, management and treatment response. Abstract Background: Acute myeloid leukemia (AML) and chronic myeloid leukemia (CML) represent a group of hematological malignancies characterized by the pathogenic clonal expansion of leukemic myeloid cells. The diagnosis and clinical outcome of AML and CML are complicated by genetic heterogeneity of disease; therefore, the identification of novel molecular biomarkers and pharmacological targets is of paramount importance. Methods: RNA-seq-based transcriptome data from a total of five studies were extracted from NCBI GEO repository and subjected to an in-depth bioinformatics analysis to identify differentially expressed genes (DEGs) between AML and CML. A systemic literature survey and functional gene ontology (GO) enrichment analysis were performed for the top 100 DEGs to identify novel candidate genes and biological processes associated with AML and CML. Results: LINC01554, PTMAP12, LOC644936, RPS27AP20 and FAM133CP were identified as novel risk genes for AML and CML. GO enrichment analysis showed that DEGs were significantly associated with pre-RNA splicing, reactive oxygen species and glycoprotein metabolism, the cellular endomembrane system, neutrophil migration and antimicrobial immune response. Conclusions: Our study revealed novel biomarkers and specific biological processes associated with AML and CML. Further studies are required to evaluate their value as molecular targets for managing and treating the myeloid malignancies.
Collapse
Affiliation(s)
- Staša Jurgec
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Laboratory for Biochemistry, Molecular Biology and Genomics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Gregor Jezernik
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Mario Gorenjak
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Tomaž Büdefeld
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Uroš Potočnik
- Center for Human Genetics and Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Laboratory for Biochemistry, Molecular Biology and Genomics, Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
- Department for Science and Research, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia
- Correspondence: ; Tel.: +386-2-2345-854
| |
Collapse
|
26
|
Computational gene expression analysis reveals distinct molecular subgroups of T-cell prolymphocytic leukemia. PLoS One 2022; 17:e0274463. [PMID: 36129940 PMCID: PMC9491575 DOI: 10.1371/journal.pone.0274463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/29/2022] [Indexed: 11/20/2022] Open
Abstract
T-cell prolymphocytic leukemia (T-PLL) is a rare blood cancer with poor prognosis. Overexpression of the proto-oncogene TCL1A and missense mutations of the tumor suppressor ATM are putative main drivers of T-PLL development, but so far only little is known about the existence of T-PLL gene expression subtypes. We performed an in-depth computational reanalysis of 68 gene expression profiles of one of the largest currently existing T-PLL patient cohorts. Hierarchical clustering combined with bootstrapping revealed three robust T-PLL gene expression subgroups. Additional comparative analyses revealed similarities and differences of these subgroups at the level of individual genes, signaling and metabolic pathways, and associated gene regulatory networks. Differences were mainly reflected at the transcriptomic level, whereas gene copy number profiles of the three subgroups were much more similar to each other, except for few characteristic differences like duplications of parts of the chromosomes 7, 8, 14, and 22. At the network level, most of the 41 predicted potential major regulators showed subgroup-specific expression levels that differed at least in comparison to one other subgroup. Functional annotations suggest that these regulators contribute to differences between the subgroups by altering processes like immune responses, angiogenesis, cellular respiration, cell proliferation, apoptosis, or migration. Most of these regulators are known from other cancers and several of them have been reported in relation to leukemia (e.g. AHSP, CXCL8, CXCR2, ELANE, FFAR2, G0S2, GIMAP2, IL1RN, LCN2, MBTD1, PPP1R15A). The existence of the three revealed T-PLL subgroups was further validated by a classification of T-PLL patients from two other smaller cohorts. Overall, our study contributes to an improved stratification of T-PLL and the observed subgroup-specific molecular characteristics could help to develop urgently needed targeted treatment strategies.
Collapse
|
27
|
Bizymi N, Matthaiou AM, Matheakakis A, Voulgari I, Aresti N, Zavitsanou K, Karasachinidis A, Mavroudi I, Pontikoglou C, Papadaki HA. New Perspectives on Myeloid-Derived Suppressor Cells and Their Emerging Role in Haematology. J Clin Med 2022; 11:jcm11185326. [PMID: 36142973 PMCID: PMC9504532 DOI: 10.3390/jcm11185326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature cells of myeloid origin that have gained researchers’ attention, as they constitute promising biomarkers and targets for novel therapeutic strategies (i.e., blockage of development, differentiation, depletion, and deactivation) in several conditions, including neoplastic, autoimmune, infective, and inflammatory diseases, as well as pregnancy, obesity, and graft rejection. They are characterised in humans by the typical immunophenotype of CD11b+CD33+HLA-DR–/low and immune-modulating properties leading to decreased T-cell proliferation, induction of T-regulatory cells (T-regs), hindering of natural killer (NK) cell functionality, and macrophage M2-polarisation. The research in the field is challenging, as there are still difficulties in defining cell-surface markers and gating strategies that uniquely identify the different populations of MDSCs, and the currently available functional assays are highly demanding. There is evidence that MDSCs display altered frequency and/or functionality and could be targeted in immune-mediated and malignant haematologic diseases, although there is a large variability of techniques and results between different laboratories. This review presents the current literature concerning MDSCs in a clinical point of view in an attempt to trigger future investigation by serving as a guide to the clinical haematologist in order to apply them in the context of precision medicine as well as the researcher in the field of experimental haematology.
Collapse
Affiliation(s)
- Nikoleta Bizymi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Andreas M. Matthaiou
- Laboratory of Molecular and Cellular Pneumonology, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Respiratory Physiology Laboratory, Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Angelos Matheakakis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Ioanna Voulgari
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Nikoletta Aresti
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Konstantina Zavitsanou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Anastasios Karasachinidis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Irene Mavroudi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Charalampos Pontikoglou
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Helen A. Papadaki
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Correspondence: ; Tel.: +30-2810394637
| |
Collapse
|
28
|
[Relapse mechanism and coping strategies of CD19 chimeric antigen receptor T cells in the treatment of diffuse large B-cell lymphoma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:788-792. [PMID: 36709176 PMCID: PMC9613498 DOI: 10.3760/cma.j.issn.0253-2727.2022.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Yu S, Ren X, Li L. Myeloid-derived suppressor cells in hematologic malignancies: two sides of the same coin. Exp Hematol Oncol 2022; 11:43. [PMID: 35854339 PMCID: PMC9295421 DOI: 10.1186/s40164-022-00296-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 07/13/2022] [Indexed: 12/15/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of bone marrow cells originating from immature myeloid cells. They exert potent immunosuppressive activity and are closely associated with the development of various diseases such as malignancies, infections, and inflammation. In malignant tumors, MDSCs, one of the most dominant cellular components comprising the tumor microenvironment, play a crucial role in tumor growth, drug resistance, recurrence, and immune escape. Although the role of MDSCs in solid tumors is currently being extensively studied, little is known about their role in hematologic malignancies. In this review, we comprehensively summarized and reviewed the different roles of MDSCs in hematologic malignancies and hematopoietic stem cell transplantation, and finally discussed current targeted therapeutic strategies.Affiliation: Kindly check and confirm the processed affiliations are correct. Amend if any.correct
Collapse
Affiliation(s)
- Shunjie Yu
- Department of Hematology, Tianjin Medical University General Hospital, Heping district 154 Anshan Road, Tianjin, China
| | - Xiaotong Ren
- Department of Hematology, Tianjin Medical University General Hospital, Heping district 154 Anshan Road, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Heping district 154 Anshan Road, Tianjin, China.
| |
Collapse
|
30
|
Cioccarelli C, Molon B. MDSCs and T cells in solid tumors and non-Hodgkin lymphomas: an immunosuppressive speech. Clin Exp Immunol 2022; 208:147-157. [PMID: 35348617 PMCID: PMC9188344 DOI: 10.1093/cei/uxac025] [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: 01/12/2022] [Accepted: 03/07/2022] [Indexed: 12/14/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous subset of cells expanded during multiple pathological settings, including cancers. In tumors, MDSCs are dominant drivers of T-cell immunosuppression. To accomplish their job, they exploit multiple mechanisms ultimately leading to the paralysis of anti-tumor immunity. Among the variety of MDSC-ways of working within the tumor microenvironment, the generation of reactive species and the metabolic reprogramming have emerged as pivotal determinants of their immunosuppressive power. In this review we will overview integral mechanisms of MDSC-mediated immunosuppression in solid tumors, with a particular focus on Non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Chiara Cioccarelli
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Italy
| | - Barbara Molon
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, Padova, Italy
| |
Collapse
|
31
|
Wang Y, Wang J, Zhu F, Wang H, Yi L, Huang K, Zhai Z. Elevated circulating myeloid-derived suppressor cells associated with poor prognosis in B-cell non-Hodgkin's lymphoma patients. Immun Inflamm Dis 2022; 10:e616. [PMID: 35478441 PMCID: PMC9017625 DOI: 10.1002/iid3.616] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Myeloid‐derived suppressor cells (MDSCs) are a heterogeneous cell population with the ability to suppress immune responses. MDSCs usually cluster in cancer, inflammation, and autoimmune diseases. Although there have been some studies on MDSCs in non‐Hodgkin lymphoma (NHL), the correlation between the peripheral levels of MDSCs in patients with various subtypes of B cell NHL and clinical features and prognosis remains inconclusive. This study aimed at the issue. Methods 101 patients with B cell NHL and 15 age‐matched healthy controls were included in this study. Flow cytometric detection of monocytic‐MDSCs (M‐MDSCs) and granulocytic‐MDSCs (G‐MDSCs) was done. Results In this study, we found that counts of circulating M‐MDSCs and G‐MDSCs were significantly increased in different clinical statuses of B‐NHL patients compared to healthy controls. Similarly, a significant increase in the levels of M‐MDSCs and G‐MDSCs was found among the diverse types of B‐NHL compared with healthy donors. Stratification studies indicated MDSCs expansion was closely associated with disease progression (tumor stage, LDH levels and B syndromes). Moreover, the overall survival time of patients with G‐MDSCs (%) ≥ 98.70% was shorter than patients with G‐MDSCs (%) < 98.70% in newly diagnosed B‐NHL subgroup, meanwhile, there was a significant difference in survival of patients with M‐MDSCs (%) ≥ 7.19% compared to patients with M‐MDSCs (%) < 7.19% in relapsed B‐NHL subgroup. Conclusion Our results suggested that M‐MDSCs and G‐MDSCs may be a potential and efficient index to evaluate the prognosis of B‐NHL patients.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jiyu Wang
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fengfeng Zhu
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Huiping Wang
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Liuying Yi
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Keke Huang
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhimin Zhai
- Department of Hematology, Second Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
32
|
Li M, Yu H, Qi F, Ye Y, Hu D, Cao J, Wang D, Mi L, Wang Z, Ding N, Ping L, Shu S, Zhu J. Anti-CD47 immunotherapy in combination with BCL-2 inhibitor to enhance anti-tumor activity in B-cell lymphoma. Hematol Oncol 2022; 40:596-608. [PMID: 35477179 DOI: 10.1002/hon.3009] [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: 01/03/2022] [Revised: 04/01/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022]
Abstract
CD47 expressed on cancer cells enables macrophage immune evasion. Blocking CD47 using anti-CD47 monoclonal antibodies (mAbs) is a promising strategy. The anti-CD47 mAb TJC4 has anti-tumor activity but lacks hematological toxicity. Venetoclax, a B-cell lymphoma 2 (BCL-2) inhibitor for B-cell malignancy, induces phosphatidylserine (PS) extracellular exposure, representing an 'eat-me' signal for macrophages. The present study aimed to explore whether TJC4-Venetoclax combined therapy exerts synergistic anti-cancer properties in B-cell lymphoma. In vitro, flow cytometry and microscopy assessed whether TJC4 monotherapy or combination treatment could promote macrophage-mediated phagocytosis of tumor cells. Induced PS exposure on the cell membrane was measured using flow cytometry with Annexin V-FITC staining. In vivo, Venetoclax and TJC4's synergistic anti-tumor effects were evaluated. B cell lymphoma cell lines express high levels of CD47 and patients with diffuse large B cell lymphoma expressing CD47 have a worse clinical prognosis. TJC4 eliminates tumor cells via macrophage-mediated phagocytosis. In vitro and in vivo, the TJC4-Venetoclax combination increased phagocytosis significantly compared with either agent alone, showing synergistic phagocytosis, and displayed synergistic anti-cancer properties in B-cell lymphoma. Our results support the TJC4-Venetoclax combination as a promising therapy, and suppressing BCL-2 and CD47 simultaneously could represent a novel therapeutic paradigm for B-cell lymphoma. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Miaomiao Li
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Hui Yu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Fei Qi
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Yingying Ye
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Dingyao Hu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Jiaowu Cao
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Dedao Wang
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Lan Mi
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | | | - Ning Ding
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Lingyan Ping
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| | - Shaokun Shu
- Department of Biomedical Engineering, Peking University, Beijing, 100871, China
| | - Jun Zhu
- Department of Lymphoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute. Beijing, 100142, China
| |
Collapse
|
33
|
Grazioli P, Orlando A, Giordano N, Noce C, Peruzzi G, Abdollahzadeh B, Screpanti I, Campese AF. Notch-Signaling Deregulation Induces Myeloid-Derived Suppressor Cells in T-Cell Acute Lymphoblastic Leukemia. Front Immunol 2022; 13:809261. [PMID: 35444651 PMCID: PMC9013886 DOI: 10.3389/fimmu.2022.809261] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/09/2022] [Indexed: 12/28/2022] Open
Abstract
Notch receptors deeply influence T-cell development and differentiation, and their dysregulation represents a frequent causative event in "T-cell acute lymphoblastic leukemia" (T-ALL). "Myeloid-derived suppressor cells" (MDSCs) inhibit host immune responses in the tumor environment, favoring cancer progression, as reported in solid and hematologic tumors, with the notable exception of T-ALL. Here, we prove that Notch-signaling deregulation in immature T cells promotes CD11b+Gr-1+ MDSCs in the Notch3-transgenic murine model of T-ALL. Indeed, aberrant T cells from these mice can induce MDSCs in vitro, as well as in immunodeficient hosts. Conversely, anti-Gr1-mediated depletion of MDSCs in T-ALL-bearing mice reduces proliferation and expansion of malignant T cells. Interestingly, the coculture with Notch-dependent T-ALL cell lines, sustains the induction of human CD14+HLA-DRlow/neg MDSCs from healthy-donor PBMCs that are impaired upon exposure to gamma-secretase inhibitors. Notch-independent T-ALL cells do not induce MDSCs, suggesting that Notch-signaling activation is crucial for this process. Finally, in both murine and human models, IL-6 mediates MDSC induction, which is significantly reversed by treatment with neutralizing antibodies. Overall, our results unveil a novel role of Notch-deregulated T cells in modifying the T-ALL environment and represent a strong premise for the clinical assessment of MDSCs in T-ALL patients.
Collapse
Affiliation(s)
- Paola Grazioli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Andrea Orlando
- Department of Molecular Medicine, Sapienza University, Rome, Italy.,Center for Life Nano- and Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Nike Giordano
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Claudia Noce
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano- and Neuro-Science, Fondazione Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | | | | | | |
Collapse
|
34
|
Tumor Immune Microenvironment in Lymphoma: Focus on Epigenetics. Cancers (Basel) 2022; 14:cancers14061469. [PMID: 35326620 PMCID: PMC8946119 DOI: 10.3390/cancers14061469] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
Lymphoma is a neoplasm arising from B or T lymphocytes or natural killer cells characterized by clonal lymphoproliferation. This tumor comprises a diverse and heterogeneous group of malignancies with distinct clinical, histopathological, and molecular characteristics. Despite advances in lymphoma treatment, clinical outcomes of patients with relapsed or refractory disease remain poor. Thus, a deeper understanding of molecular pathogenesis and tumor progression of lymphoma is required. Epigenetic alterations contribute to cancer initiation, progression, and drug resistance. In fact, over the past decade, dysregulation of epigenetic mechanisms has been identified in lymphomas, and the knowledge of the epigenetic aberrations has led to the emergence of the promising epigenetic therapy field in lymphoma tumors. However, epigenetic aberrations in lymphoma not only have been found in tumor cells, but also in cells from the tumor microenvironment, such as immune cells. Whereas the epigenetic dysregulation in lymphoma cells is being intensively investigated, there are limited studies regarding the epigenetic mechanisms that affect the functions of immune cells from the tumor microenvironment in lymphoma. Therefore, this review tries to provide a general overview of epigenetic alterations that affect both lymphoma cells and infiltrating immune cells within the tumor, as well as the epigenetic cross-talk between them.
Collapse
|
35
|
Single-cell RNA sequencing reveals evolution of immune landscape during glioblastoma progression. Nat Immunol 2022; 23:971-984. [PMID: 35624211 PMCID: PMC9174057 DOI: 10.1038/s41590-022-01215-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 04/18/2022] [Indexed: 01/22/2023]
Abstract
Glioblastoma (GBM) is an incurable primary malignant brain cancer hallmarked with a substantial protumorigenic immune component. Knowledge of the GBM immune microenvironment during tumor evolution and standard of care treatments is limited. Using single-cell transcriptomics and flow cytometry, we unveiled large-scale comprehensive longitudinal changes in immune cell composition throughout tumor progression in an epidermal growth factor receptor-driven genetic mouse GBM model. We identified subsets of proinflammatory microglia in developing GBMs and anti-inflammatory macrophages and protumorigenic myeloid-derived suppressors cells in end-stage tumors, an evolution that parallels breakdown of the blood-brain barrier and extensive growth of epidermal growth factor receptor+ GBM cells. A similar relationship was found between microglia and macrophages in patient biopsies of low-grade glioma and GBM. Temozolomide decreased the accumulation of myeloid-derived suppressor cells, whereas concomitant temozolomide irradiation increased intratumoral GranzymeB+ CD8+T cells but also increased CD4+ regulatory T cells. These results provide a comprehensive and unbiased immune cellular landscape and its evolutionary changes during GBM progression.
Collapse
|
36
|
Guha P, Katz SC. Strategies for manufacturing cell therapy products aligned with patient needs. Methods Cell Biol 2022; 167:203-226. [DOI: 10.1016/bs.mcb.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
37
|
Zhang Z, Kuo JCT, Yao S, Zhang C, Khan H, Lee RJ. CpG Oligodeoxynucleotides for Anticancer Monotherapy from Preclinical Stages to Clinical Trials. Pharmaceutics 2021; 14:73. [PMID: 35056969 PMCID: PMC8780291 DOI: 10.3390/pharmaceutics14010073] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/16/2021] [Accepted: 12/26/2021] [Indexed: 11/29/2022] Open
Abstract
CpG oligodeoxynucleotides (CpG ODNs), the artificial versions of unmethylated CpG motifs that were originally discovered in bacterial DNA, are demonstrated not only as potent immunoadjuvants but also as anticancer agents by triggering toll-like receptor 9 (TLR9) activation in immune cells. TLR9 activation triggered by CpG ODN has been shown to activate plasmacytoid dendritic cells (pDCs) and cytotoxic T lymphocytes (CTLs), enhancing T cell-mediated antitumor immunity. However, the extent of antitumor immunity carried by TLR agonists has not been optimized individually or in combinations with cancer vaccines, resulting in a decreased preference for TLR agonists as adjuvants in clinical trials. Although various combination therapies involving CpG ODNs have been applied in clinical trials, none of the CpG ODN-based drugs have been approved by the FDA, owing to the short half-life of CpG ODNs in serum that leads to low activation of natural killer cells (NK cells) and CTLs, along with increases of pro-inflammatory cytokine productions. This review summarized the current innovation on CpG ODNs that are under clinical investigation and explored the future direction for CpG ODN-based nanomedicine as an anticancer monotherapy.
Collapse
Affiliation(s)
- Zhongkun Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH 43210, USA; (Z.Z.); (J.C.-T.K.); (C.Z.); (H.K.)
| | - Jimmy Chun-Tien Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH 43210, USA; (Z.Z.); (J.C.-T.K.); (C.Z.); (H.K.)
| | - Siyu Yao
- Department of Food Science and Technology, The Ohio State University, 110 Parker Food Science and Technology Building, 2015 Fyffe Road, Columbus, OH 43210, USA;
| | - Chi Zhang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH 43210, USA; (Z.Z.); (J.C.-T.K.); (C.Z.); (H.K.)
| | - Hira Khan
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH 43210, USA; (Z.Z.); (J.C.-T.K.); (C.Z.); (H.K.)
- Department of Pharmacy, Abbottabad University of Science and Technology, Havelian, Abbottabad 22500, Pakistan
| | - Robert J. Lee
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, 500 W 12th Avenue, Columbus, OH 43210, USA; (Z.Z.); (J.C.-T.K.); (C.Z.); (H.K.)
| |
Collapse
|
38
|
Huang Q, Lin J, Huang S, Shen J. Impact of Qi-Invigorating Traditional Chinese Medicines on Diffuse Large B Cell Lymphoma Based on Network Pharmacology and Experimental Validation. Front Pharmacol 2021; 12:787816. [PMID: 34955857 PMCID: PMC8699731 DOI: 10.3389/fphar.2021.787816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/15/2021] [Indexed: 12/31/2022] Open
Abstract
Background: It has been verified that deficiency of Qi, a fundamental substance supporting daily activities according to the Traditional Chinese Medicine theory, is an important symptom of cancer. Qi-invigorating herbs can inhibit cancer development through promoting apoptosis and improving cancer microenvironment. In this study, we explored the potential mechanisms of Qi-invigorating herbs in diffuse large B cell lymphoma (DLBCL) through network pharmacology and in vitro experiment. Methods: Active ingredients of Qi-invigorating herbs were predicted from the Traditional Chinese Medicine Systems Pharmacology Database. Potential targets were obtained via the SwissTargetPrediction and STITCH databases. Target genes of DLBCL were obtained through the PubMed, the gene-disease associations and the Malacards databases. Overlapping genes between DLBCL and each Qi-invigorating herb were collected. Hub genes were subsequently screened via Cytoscape. The Gene Ontology and pathway enrichment analyses were performed using the DAVID database. Molecular docking was performed among active ingredients and hub genes. Hub genes linked with survival and tumor microenvironment were analyzed through the GEPIA 2.0 and TIMER 2.0 databases, respectively. Additionally, in vitro experiment was performed to verify the roles of common hub genes. Results: Through data mining, 14, 4, 22, 22, 35, 2, 36 genes were filtered as targets of Ginseng Radix et Rhizoma, Panacis Quinquefolii Radix, Codonopsis Radix, Pseudostellariae Radix, Astragali Radix, Dioscoreae Rhizoma, Glycyrrhizae Radix et Rhizoma for DLBCL treatment, respectively. Then besides Panacis Quinquefolii Radix and Dioscoreae Rhizoma, 1,14, 10, 14,13 hub genes were selected, respectively. Molecular docking studies indicated that active ingredients could stably bind to the pockets of hub proteins. CASP3, CDK1, AKT1 and MAPK3 were predicted as common hub genes. However, through experimental verification, only CASP3 was considered as the common target of Qi-invigorating herbs on DLBCL apoptosis. Furthermore, the TIMER2.0 database showed that Qi-invigorating herbs might act on DLBCL microenvironment through their target genes. Tumor-associated neutrophils may be main target cells of DLBCL treated by Qi-invigorating herbs. Conclusion: Our results support the effects of Qi-invigorating herbs on DLBCL. Hub genes and immune infiltrating cells provided the molecular basis for each Qi-invigorating herb acting on DLBCL.
Collapse
Affiliation(s)
- Qian Huang
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jinkun Lin
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Surong Huang
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jianzhen Shen
- Fujian Institute of Hematology, Fujian Medical Center of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
39
|
Le Gallou S, Lhomme F, Irish JM, Mingam A, Pangault C, Monvoisin C, Ferrant J, Azzaoui I, Rossille D, Bouabdallah K, Damaj G, Cartron G, Godmer P, Le Gouill S, Casasnovas RO, Molina TJ, Houot R, Lamy T, Tarte K, Fest T, Roussel M. Nonclassical Monocytes Are Prone to Migrate Into Tumor in Diffuse Large B-Cell Lymphoma. Front Immunol 2021; 12:755623. [PMID: 34975843 PMCID: PMC8716558 DOI: 10.3389/fimmu.2021.755623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022] Open
Abstract
Absolute count of circulating monocytes has been proposed as an independent prognostic factor in diffuse large B-cell lymphoma (DLBCL). However, monocyte nomenclature includes various subsets with pro-, anti-inflammatory, or suppressive functions, and their clinical relevance in DLBCL has been poorly explored. Herein, we broadly assessed circulating monocyte heterogeneity in 91 DLBCL patients. Classical- (cMO, CD14pos CD16neg) and intermediate- (iMO, CD14pos CD16pos) monocytes accumulated in DLBCL peripheral blood and exhibited an inflammatory phenotype. On the opposite, nonclassical monocytes (ncMOSlanpos, CD14low CD16pos Slanneg and ncMOSlanneg, CD14low CD16pos, Slanneg) were decreased in peripheral blood. Tumor-conditioned monocytes presented similarities with ncMO phenotype from DLBCL and were prone to migrate in response to CCL5 and CXCL12, and presented similarities with DLBCL-infiltrated myeloid cells, as defined by mass cytometry. Finally, we demonstrated the adverse value of an accumulation of nonclassical monocytes in 2 independent cohorts of DLBCL.
Collapse
Affiliation(s)
- Simon Le Gallou
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Faustine Lhomme
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Jonathan M. Irish
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Anna Mingam
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
| | - Celine Pangault
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Celine Monvoisin
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Juliette Ferrant
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Imane Azzaoui
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
| | - Delphine Rossille
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Krimo Bouabdallah
- Centre Hospitalier Universitaire de Bordeaux, Service d’Hématologie Clinique, Bordeaux, France
| | - Gandhi Damaj
- Centre Hospitalier Universitaire de Caen, Service d’Hématologie Clinique, Caen, France
| | - Guillaume Cartron
- Centre Hospitalier Universitaire de Montpellier, Service d’Hématologie Clinique, Montpellier, France
| | - Pascal Godmer
- Centre Hospitalier de Bretagne Atlantique, Unité d’Hématologie Clinique, Vannes, France
| | - Steven Le Gouill
- Centre Hospitalier Universitaire de Nantes, Service d’Hématologie Clinique, Institut National de la Sante et de la Recherche Medicale, Centre de Recherche en Cancérologie et Immunologie Nantes Angers (INSERM CCRCINA) Nantes-Angers, NeXT Université de Nantes, Nantes, France
| | | | - Thierry Jo Molina
- Asistance Publique, Hopitaux de Paris (APHP), Necker, Service d’Anatomopathologie, Sorbonne Université, Paris, France
| | - Roch Houot
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
- Centre Hospitalier Universitaire de Rennes, Service d’Hématologie Clinique, Rennes, France
| | - Thierry Lamy
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
- Centre Hospitalier Universitaire de Rennes, Service d’Hématologie Clinique, Rennes, France
| | - Karin Tarte
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Thierry Fest
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| | - Mikael Roussel
- Centre Hospitalier Universitaire de Rennes, Pôle Biologie, Rennes, France
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche U1236, Université Rennes 1, Etablissement Français du Sang Bretagne, Rennes, France
| |
Collapse
|
40
|
Human CD22-Transgenic, Primary Murine Lymphoma Challenges Immunotherapies in Organ-Specific Tumor Microenvironments. Int J Mol Sci 2021; 22:ijms221910433. [PMID: 34638774 PMCID: PMC8508822 DOI: 10.3390/ijms221910433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022] Open
Abstract
Targeted immunotherapies have greatly changed treatment of patients with B cell malignancies. To further enhance immunotherapies, research increasingly focuses on the tumor microenvironment (TME), which differs considerably by organ site. However, immunocompetent mouse models of disease to study immunotherapies targeting human molecules within organ-specific TME are surprisingly rare. We developed a myc-driven, primary murine lymphoma model expressing a human-mouse chimeric CD22 (h/mCD22). Stable engraftment of three distinct h/mCD22+ lymphoma was established after subcutaneous and systemic injection. However, only systemic lymphoma showed immune infiltration that reflected human disease. In this model, myeloid cells supported lymphoma growth and showed a phenotype of myeloid-derived suppressor cells. The human CD22-targeted immunotoxin Moxetumomab was highly active against h/mCD22+ lymphoma and similarly reduced infiltration of bone marrow and spleen of all three models up to 90-fold while efficacy against lymphoma in lymph nodes varied substantially, highlighting relevance of organ-specific TME. As in human aggressive lymphoma, anti-PD-L1 as monotherapy was not efficient. However, anti-PD-L1 enhanced efficacy of Moxetumomab suggesting potential for future clinical application. The novel model system of h/mCD22+ lymphoma provides a unique platform to test targeted immunotherapies and may be amenable for other human B cell targets such as CD19 and CD20.
Collapse
|
41
|
Liu Y, Zhou X, Wang X. Targeting the tumor microenvironment in B-cell lymphoma: challenges and opportunities. J Hematol Oncol 2021; 14:125. [PMID: 34404434 PMCID: PMC8369706 DOI: 10.1186/s13045-021-01134-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
B-cell lymphoma is a group of hematological malignancies with high clinical and biological heterogeneity. The pathogenesis of B-cell lymphoma involves a complex interaction between tumor cells and the tumor microenvironment (TME), which is composed of stromal cells and extracellular matrix. Although the roles of the TME have not been fully elucidated, accumulating evidence implies that TME is closely relevant to the origination, invasion and metastasis of B-cell lymphoma. Explorations of the TME provide distinctive insights for cancer therapy. Here, we epitomize the recent advances of TME in B-cell lymphoma and discuss its function in tumor progression and immune escape. In addition, the potential clinical value of targeting TME in B-cell lymphoma is highlighted, which is expected to pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Yingyue Liu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, No. 324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
42
|
Papafragkos I, Markaki E, Kalpadakis C, Verginis P. Decoding the Myeloid-Derived Suppressor Cells in Lymphoid Malignancies. J Clin Med 2021; 10:jcm10163462. [PMID: 34441758 PMCID: PMC8397155 DOI: 10.3390/jcm10163462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are immature myeloid precursors which emerged as a potent regulator of the immune system, exerting suppressive properties in diverse disease settings. In regards to cancer, MDSCs have an established role in solid tumors; however, their contribution to immune regulation during hematologic malignancies and particularly in lymphomas remains ill-defined. Herein focused on lymphoma, we discuss the literature on MDSC cells in all histologic types, and we also refer to lessons learned by animal models of lymphoma. Furthermore, we elaborate on future directions and unmet needs and challenges in the MDSC field related to lymphoma malignancies which may shed light on the complex nature of the immune system in malignancies.
Collapse
Affiliation(s)
- Iosif Papafragkos
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, 71003 Heraklion, Greece; (I.P.); (E.M.)
| | - Efrosyni Markaki
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, 71003 Heraklion, Greece; (I.P.); (E.M.)
| | - Christina Kalpadakis
- Laboratory of Haematology, Division of Laboratory Medicine, Medical School, University of Crete, 71003 Heraklion, Greece
- Department of Laboratory Haematology, University Hospital of Heraklion, 71500 Heraklion, Greece
- Correspondence: (C.K.); (P.V.); Tel.: +30-69-4458-2738 (C.K.); +30-28-1039-4553 (P.V.)
| | - Panayotis Verginis
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, 71003 Heraklion, Greece; (I.P.); (E.M.)
- Department of Laboratory Haematology, University Hospital of Heraklion, 71500 Heraklion, Greece
- Correspondence: (C.K.); (P.V.); Tel.: +30-69-4458-2738 (C.K.); +30-28-1039-4553 (P.V.)
| |
Collapse
|
43
|
Vito A, Salem O, El-Sayes N, MacFawn IP, Portillo AL, Milne K, Harrington D, Ashkar AA, Wan Y, Workenhe ST, Nelson BH, Bruno TC, Mossman KL. Immune checkpoint blockade in triple negative breast cancer influenced by B cells through myeloid-derived suppressor cells. Commun Biol 2021; 4:859. [PMID: 34253827 PMCID: PMC8275624 DOI: 10.1038/s42003-021-02375-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022] Open
Abstract
Triple negative breast cancer holds a dismal clinical outcome and as such, patients routinely undergo aggressive, highly toxic treatment regimens. Clinical trials for TNBC employing immune checkpoint blockade in combination with chemotherapy show modest prognostic benefit, but the percentage of patients that respond to treatment is low, and patients often succumb to relapsed disease. Here, we show that a combination immunotherapy platform utilizing low dose chemotherapy (FEC) combined with oncolytic virotherapy (oHSV-1) increases tumor-infiltrating lymphocytes, in otherwise immune-bare tumors, allowing 60% of mice to achieve durable tumor regression when treated with immune checkpoint blockade. Whole-tumor RNA sequencing of mice treated with FEC + oHSV-1 shows an upregulation of B cell receptor signaling pathways and depletion of B cells prior to the start of treatment in mice results in complete loss of therapeutic efficacy and expansion of myeloid-derived suppressor cells. Additionally, RNA sequencing data shows that FEC + oHSV-1 suppresses genes associated with myeloid-derived suppressor cells, a key population of cells that drive immune escape and mediate therapeutic resistance. These findings highlight the importance of tumor-infiltrating B cells as drivers of antitumor immunity and their potential role in the regulation of myeloid-derived suppressor cells.
Collapse
Affiliation(s)
- Alyssa Vito
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Omar Salem
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Nader El-Sayes
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ian P MacFawn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ana L Portillo
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Katy Milne
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | | | - Ali A Ashkar
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Yonghong Wan
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Samuel T Workenhe
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Karen L Mossman
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
- Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
44
|
Popovic LS, Matovina-Brko G, Popovic M, Popovic M, Cvetanovic A, Nikolic I, Kukic B, Petrovic D. Immunotherapy in the treatment of lymphoma. World J Stem Cells 2021; 13:503-520. [PMID: 34249225 PMCID: PMC8246244 DOI: 10.4252/wjsc.v13.i6.503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/31/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023] Open
Abstract
Relapsed or refractory non-Hodgkin’s lymphomas, especially diffuse large B-cell lymphoma as well as relapsed or refractory Hodgkin lymphomas are hard-to-treat diseases. Patients who do not respond to initial therapy or experience relapse are treated with salvage regimens, and if eligible for aggressive therapy, treatment is continued with high-dose chemotherapy and autologous stem cell transplantation. Current therapy options can cure substantial numbers of patients, however for some it is still an uncurable disease. Numerous new drugs and cell therapies are being investigated for the treatment of relapsed or refractory lymphomas. Different types of immunotherapy options have shown promising results, and some have already become the standard of care. Here, we review immunotherapy options for the treatment of lymphoma and discuss the results, positions, practical aspects, and future directions of different drugs and cellular therapies for the treatment of this disease.
Collapse
Affiliation(s)
- Lazar S Popovic
- Department for Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Novi Sad 21000, Serbia
| | - Gorana Matovina-Brko
- Department for Medical Oncology, Oncology Institute of Vojvodina, Novi Sad 21000, Serbia
| | - Maja Popovic
- Department for Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Novi Sad 21000, Serbia
| | - Milica Popovic
- Department for Nephrology and Clinical Immunology, Clinical Center of Vojvodina, University of Novi Sad, Novi Sad 21000, Serbia
| | - Ana Cvetanovic
- Department for Medical Oncology, Clinical Center of Nis, University of Nis, Nis 18000, Serbia
| | - Ivan Nikolic
- Department for Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Novi Sad 21000, Serbia
| | - Biljana Kukic
- Department for Medical Oncology, Oncology Institute of Vojvodina, University of Novi Sad, Novi Sad 21000, Serbia
| | - Dragana Petrovic
- Department for Medical Oncology, Oncology Institute of Vojvodina, Novi Sad 21000, Serbia
| |
Collapse
|
45
|
Jiménez-Cortegana C, Palazón-Carrión N, Martin Garcia-Sancho A, Nogales-Fernandez E, Carnicero-González F, Ríos-Herranz E, de la Cruz-Vicente F, Rodríguez-García G, Fernández-Álvarez R, Rueda Dominguez A, Casanova-Espinosa M, Martínez-Banaclocha N, Gumà-Padrò J, Gómez-Codina J, Labrador J, Salar-Silvestre A, Rodriguez-Abreu D, Galvez-Carvajal L, Provencio M, Sánchez-Beato M, Guirado-Risueño M, Espejo-García P, Lejeune M, Álvaro T, Sánchez-Margalet V, de la Cruz-Merino L. Circulating myeloid-derived suppressor cells and regulatory T cells as immunological biomarkers in refractory/relapsed diffuse large B-cell lymphoma: translational results from the R2-GDP-GOTEL trial. J Immunother Cancer 2021; 9:jitc-2020-002323. [PMID: 34158317 PMCID: PMC8728348 DOI: 10.1136/jitc-2020-002323] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2021] [Indexed: 12/18/2022] Open
Abstract
Background The search for immunological markers with ability of predicting clinical outcome is a priority in lymphomas, and in cancer in general. It is well known that some immunomodulatory cells, such as myeloid derived suppressor cells (MDSCs) or regulatory T cells (Tregs), are recruited by tumors, jeopardizing antitumor immunosurveillance. In this work, we have studied blood levels of these immunosuppressive cells in patients with relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL), prior to and along the course of the experimental rituximab, gemcitabine, dexamethasone, and cisplatin (R2-GDP) schedule, as a translational substudy of the R2-GDP-GOTEL trial (EudraCT Number: 2014-001620-29), which included lenalidomide as an immunomodulator. Methods Blood samples were taken before treatment, at cycle 3 and end of induction. Samples were analyzed by flow cytometry. Non-parametric tests were used. Mann-Whitney U test was used to compare basal cells distributions, and Wilcoxon test was considered to compare cells distribution at different times. Spearman test was performed to measure the degree of association between cell populations. Results In this study, MDSC and Treg circulating concentration was found increased in all patients compared with a healthy control group and decreased after treatment only in patients with longest overall survival (>24 months), reaching the levels of the healthy group. Likewise, the number of inhibited T lymphocytes expressing Programmed Death-1 (PD-1) were increased in peripheral blood from patients and decreased on the treatment, whereas activated T lymphocytes increased after therapy in those with better overall survival. Conclusions In conclusion, blood concentration of MDSCs and Treg cells may be good prognostic markers for overall survival after 2 years in R/R DLBCL. These results point to a possible role of these elements in the immunosuppression of these patients, as assessed by the circulating activated and inhibited T lymphocytes, and therefore, they may be considered as therapeutic targets in DLBCL.
Collapse
Affiliation(s)
- Carlos Jiménez-Cortegana
- Medical Biochemistry and Molecular Biology and Immunology, Virgen Macarena University Hospital, Sevilla, Seville, Spain
| | | | | | - Esteban Nogales-Fernandez
- Clinical Oncology Dept. Medicine Department, University of Seville, Virgen Macarena University Hospital, Seville, Spain
| | | | | | | | | | | | | | | | | | - Josep Gumà-Padrò
- Clinical Oncology Dept, Sant Joan de Reus University Hospital URV, IISPV, Reus, Spain
| | | | - Jorge Labrador
- Hematology Dept, Hospital Universitario de Burgos, Burgos, Spain
| | | | | | - Laura Galvez-Carvajal
- Unidad de Gestión Clínica Intercentros de Oncología Médica de Málaga, Hospitales Universitarios Regional y Virgen de la Victoria, Málaga, Spain
| | - Mariano Provencio
- Clinical Oncology Dept, Puerta de Hierro University Hospital, Majadahonda, Madrid, Spain
| | - Margarita Sánchez-Beato
- Lymphoma Research Group, Clinical Oncology Dept, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Madrid, Spain
| | - María Guirado-Risueño
- Clinical Oncology Dept, Elche General University Hospital General, Elche, Alicante, Spain
| | - Pablo Espejo-García
- Clinical Oncology Dept. Medicine Department, University of Seville, Virgen Macarena University Hospital, Seville, Spain
| | - Marylene Lejeune
- Pathology Department, Plataforma de Estudios Histológicos, Citológicos y de Digitalización, Hospital de Tortosa Verge de la Cinta, IISPV, URV, Tortosa, Tarragona, Spain
| | - Tomás Álvaro
- Pathology Dept, Hospital de Tortosa Verge de la Cinta, Catalan Institute of Health, Institut d'Investigació Sanitària Pere Virgili (IISPV), Tortosa, Tarragona, Spain
| | - Victor Sánchez-Margalet
- Medical Biochemistry and Molecular Biology and Immunology, Virgen Macarena University Hospital, Sevilla, Seville, Spain
| | | | | | | | | |
Collapse
|
46
|
Induced Pluripotent Stem Cells (iPSCs) Provide a Potentially Unlimited T Cell Source for CAR-T Cell Development and Off-the-Shelf Products. Pharm Res 2021; 38:931-945. [PMID: 34114161 DOI: 10.1007/s11095-021-03067-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/24/2021] [Indexed: 12/28/2022]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has been increasingly conducted for cancer patients in clinical settings. Progress in this therapeutic approach is hampered by the lack of a solid manufacturing process, T lymphocytes, and tumor-specific antigens. T cell source used in CAR-T cell therapy is derived predominantly from the patient's own T lymphocytes, which makes this approach impracticable to patients with progressive diseases and T leukemia. The generation of autologous CAR-T cells is time-consuming due to the lack of readily available T lymphocytes and is not applicable for third-party patients. Pluripotent stem cells, such as human induced pluripotent stem cells (hiPSCs), can provide an unlimited T cell source for CAR-T cell development with the potential of generating off-the-shelf T cell products. T-iPSCs (iPSC-derived T cells) are phenotypically defined, expandable, and as functional as physiological T cells. The combination of iPSC and CAR technologies provides an exciting opportunity to oncology and greatly facilitates cell-based therapy for cancer patients. However, T-iPSCs, in combination with CARs, are at the early stage of development and need further pre-clinical and clinical studies. This review will critically discuss the progress made in iPSC-derived T cells and provides a roadmap for the development of CAR iPSC-derived T cells and off-the-shelf T-iPSCs.
Collapse
|
47
|
Lenalidomide triggers T-cell effector functions in vivo in patients with follicular lymphoma. Blood Adv 2021; 5:2063-2074. [PMID: 33877296 DOI: 10.1182/bloodadvances.2020003774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023] Open
Abstract
The immunomodulatory drug lenalidomide is used in patients with follicular lymphoma (FL) with the aim of stimulating T-cell antitumor immune response. However, little is known about the effects of lenalidomide on T-cell biology in vivo in patients with FL. We thus undertook an extensive longitudinal immunologic study, including phenotypic, transcriptomic, and functional analyses, on 44 first-line and 27 relapsed/refractory patients enrolled in the GALEN trial (Obinutuzumab Combined With Lenalidomide for Relapsed or Refractory Follicular B-Cell Lymphoma) to test the efficacy of lenalidomide and obinutuzumab combination in patients with FL. Lenalidomide rapidly and transiently induced an activated T-cell phenotype, including HLA-DR, Tim-3, CD137, and programmed cell death protein 1 (PD-1) upregulation. Furthermore, sequential RNA-sequencing of sorted PD-1+ and PD-1- T-cell subsets revealed that lenalidomide triggered a strong enrichment for several gene signatures related to effector memory T-cell features, including proliferation, antigen receptor signaling, and immune synapse restoration; all were validated at the phenotypic level and with ex vivo functional assays. Correlative analyses pinpointed a negative clinical impact of high effector T-cell and regulatory T-cell percentages before and during treatment. Our findings bring new insight in lenalidomide mechanisms of action at work in vivo and will fuel a new rationale for the design of combination therapies.
Collapse
|
48
|
Jiménez I, Tazón-Vega B, Abrisqueta P, Nieto JC, Bobillo S, Palacio-García C, Carabia J, Valdés-Mas R, Munuera M, Puigdefàbregas L, Parra G, Esteve-Codina A, Franco-Jarava C, Iacoboni G, Terol MJ, García-Marco JA, Crespo M, Bosch F. Immunological and genetic kinetics from diagnosis to clinical progression in chronic lymphocytic leukemia. Biomark Res 2021; 9:37. [PMID: 34016160 PMCID: PMC8138982 DOI: 10.1186/s40364-021-00290-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Mechanisms driving the progression of chronic lymphocytic leukemia (CLL) from its early stages are not fully understood. The acquisition of molecular changes at the time of progression has been observed in a small fraction of patients, suggesting that CLL progression is not mainly driven by dynamic clonal evolution. In order to shed light on mechanisms that lead to CLL progression, we investigated longitudinal changes in both the genetic and immunological scenarios. METHODS We performed genetic and immunological longitudinal analysis using paired primary samples from untreated CLL patients that underwent clinical progression (sampling at diagnosis and progression) and from patients with stable disease (sampling at diagnosis and at long-term asymptomatic follow-up). RESULTS Molecular analysis showed limited and non-recurrent molecular changes at progression, indicating that clonal evolution is not the main driver of clinical progression. Our analysis of the immune kinetics found an increasingly dysfunctional CD8+ T cell compartment in progressing patients that was not observed in those patients that remained asymptomatic. Specifically, terminally exhausted effector CD8+ T cells (T-betdim/-EomeshiPD1hi) accumulated, while the the co-expression of inhibitory receptors (PD1, CD244 and CD160) increased, along with an altered gene expression profile in T cells only in those patients that progressed. In addition, malignant cells from patients at clinical progression showed enhanced capacity to induce exhaustion-related markers in CD8+ T cells ex vivo mainly through a mechanism dependent on soluble factors including IL-10. CONCLUSIONS Altogether, we demonstrate that the interaction with the immune microenvironment plays a key role in clinical progression in CLL, thereby providing a rationale for the use of early immunotherapeutic intervention.
Collapse
Affiliation(s)
- Isabel Jiménez
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Bárbara Tazón-Vega
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Pau Abrisqueta
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Juan C Nieto
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Sabela Bobillo
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Carles Palacio-García
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Júlia Carabia
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | | | - Magdalena Munuera
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Lluís Puigdefàbregas
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Genís Parra
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08003, Barcelona, Spain.,Universitat Pompeu Fabra, 08002, Barcelona, Spain
| | - Anna Esteve-Codina
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, 08003, Barcelona, Spain.,Universitat Pompeu Fabra, 08002, Barcelona, Spain
| | - Clara Franco-Jarava
- Servei d'Immunologia, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Gloria Iacoboni
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - María José Terol
- Department of Hematology, Clínic University Hospital, INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | | | - Marta Crespo
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, C/Natzaret 115-117, 08035, Barcelona, Spain.,Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Francesc Bosch
- Department de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain. .,Servei d'Hematologia, Vall d'Hebron Hospital Universitari, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
49
|
Jain MD, Zhao H, Wang X, Atkins R, Menges M, Reid K, Spitler K, Faramand R, Bachmeier C, Dean EA, Cao B, Chavez JC, Shah B, Lazaryan A, Nishihori T, Hussaini M, Gonzalez RJ, Mullinax JE, Rodriguez PC, Conejo-Garcia JR, Anasetti C, Davila ML, Locke FL. Tumor interferon signaling and suppressive myeloid cells are associated with CAR T-cell failure in large B-cell lymphoma. Blood 2021; 137:2621-2633. [PMID: 33512407 PMCID: PMC8120145 DOI: 10.1182/blood.2020007445] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/10/2020] [Indexed: 12/18/2022] Open
Abstract
Axicabtagene ciloleucel (axi-cel) is a chimeric antigen receptor (CAR) T-cell therapy for relapsed or refractory large B-cell lymphoma (LBCL). This study evaluated whether immune dysregulation, present before CAR T-cell therapy, was associated with treatment failure. Tumor expression of interferon (IFN) signaling, high blood levels of monocytic myeloid-derived suppressor cells (M-MDSCs), and high blood interleukin-6 and ferritin levels were each associated with a lack of durable response. Similar to other cancers, we found that in LBCL tumors, IFN signaling is associated with the expression of multiple checkpoint ligands, including programmed cell death-ligand 1, and these were higher in patients who lacked durable responses to CAR-T therapy. Moreover, tumor IFN signaling and blood M-MDSCs associated with decreased axi-cel expansion. Finally, patients with high tumor burden had higher immune dysregulation with increased serum inflammatory markers and tumor IFN signaling. These data support that immune dysregulation in LBCL promotes axi-cel resistance via multiple mechanistic programs: insufficient axi-cel expansion associated with both circulating M-MDSC and tumor IFN signaling, which also gives rise to expression of immune checkpoint ligands.
Collapse
Affiliation(s)
- Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| | | | | | | | | | | | | | - Rawan Faramand
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| | | | - Erin A Dean
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| | - Biwei Cao
- Department of Bioinformatics and Biostatistics
| | | | | | | | - Taiga Nishihori
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| | | | | | | | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Jose R Conejo-Garcia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, University of South Florida Morsani College of Medicine, Tampa, FL
| | - Claudio Anasetti
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| | - Marco L Davila
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| | - Frederick L Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy
| |
Collapse
|
50
|
Munari E, Mariotti FR, Quatrini L, Bertoglio P, Tumino N, Vacca P, Eccher A, Ciompi F, Brunelli M, Martignoni G, Bogina G, Moretta L. PD-1/PD-L1 in Cancer: Pathophysiological, Diagnostic and Therapeutic Aspects. Int J Mol Sci 2021; 22:5123. [PMID: 34066087 PMCID: PMC8151504 DOI: 10.3390/ijms22105123] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Immune evasion is a key strategy adopted by tumor cells to escape the immune system while promoting their survival and metastatic spreading. Indeed, several mechanisms have been developed by tumors to inhibit immune responses. PD-1 is a cell surface inhibitory receptor, which plays a major physiological role in the maintenance of peripheral tolerance. In pathological conditions, activation of the PD-1/PD-Ls signaling pathway may block immune cell activation, a mechanism exploited by tumor cells to evade the antitumor immune control. Targeting the PD-1/PD-L1 axis has represented a major breakthrough in cancer treatment. Indeed, the success of PD-1 blockade immunotherapies represents an unprecedented success in the treatment of different cancer types. To improve the therapeutic efficacy, a deeper understanding of the mechanisms regulating PD-1 expression and signaling in the tumor context is required. We provide an overview of the current knowledge of PD-1 expression on both tumor-infiltrating T and NK cells, summarizing the recent evidence on the stimuli regulating its expression. We also highlight perspectives and limitations of the role of PD-L1 expression as a predictive marker, discuss well-established and novel potential approaches to improve patient selection and clinical outcome and summarize current indications for anti-PD1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Enrico Munari
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, 25100 Brescia, Italy;
| | - Francesca R. Mariotti
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Linda Quatrini
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Pietro Bertoglio
- Division of Thoracic Surgery, IRCCS Maggiore Teaching Hospital and Sant’Orsola University Hospital, 40133 Bologna, Italy;
| | - Nicola Tumino
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Paola Vacca
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| | - Albino Eccher
- Pathology Unit, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Francesco Ciompi
- Computational Pathology Group, Department of Pathology, Radboud University Medical Center, 6543 SH Nijmegen, The Netherlands;
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (M.B.); (G.M.)
| | - Guido Martignoni
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (M.B.); (G.M.)
- Pathology Unit, Pederzoli Hospital, 37019 Peschiera del Garda, Italy
| | - Giuseppe Bogina
- Pathology Unit, IRCCS Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Lorenzo Moretta
- Immunology Area, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (F.R.M.); (L.Q.); (N.T.); (P.V.)
| |
Collapse
|