1
|
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
|
2
|
Ribatti D, Tamma R, Annese T, Ingravallo G, Specchia G. Macrophages and angiogenesis in human lymphomas. Clin Exp Med 2024; 24:26. [PMID: 38285283 PMCID: PMC10824884 DOI: 10.1007/s10238-023-01291-y] [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: 12/10/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024]
Abstract
A link exists between chronic inflammation and cancer and immune cells, angiogenesis, and tumor progression. In hematologic malignancies, tumor-associated macrophages (TAMs) are a significant part of the tumor microenvironment. Macrophages are classified into M1/classically activated and M2/alternatively activated. In tumors, TAMs are mainly constituted by M2 subtype, which promotes angiogenesis, lymphangiogenesis, repair, and remodeling, suppressing adaptive immunity, increasing tumor cell proliferation, drug resistance, histological malignancy, and poor clinical prognosis. The aim of our review article is to define the role of TAMs and their relationship with the angiogenesis in patients with lymphoma reporting both an analysis of main published data and those emerging from our studies. Finally, we have discussed the anti-angiogenic approach in the treatment of lymphomas.
Collapse
Affiliation(s)
- Domenico Ribatti
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy.
| | - Roberto Tamma
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
| | - Tiziana Annese
- Department of Translational Biomedicine and Neuroscience, University of Bari Medical School, Bari, Italy
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Medical School, Bari, Italy
| | | |
Collapse
|
3
|
Menéndez V, Solórzano JL, García-Cosío M, Alonso-Alonso R, Rodríguez M, Cereceda L, Fernández S, Díaz E, Montalbán C, Estévez M, Piris MA, García JF. Immune and stromal transcriptional patterns that influence the outcome of classic Hodgkin lymphoma. Sci Rep 2024; 14:710. [PMID: 38184757 PMCID: PMC10771441 DOI: 10.1038/s41598-024-51376-1] [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: 08/08/2023] [Accepted: 01/04/2024] [Indexed: 01/08/2024] Open
Abstract
Classic Hodgkin lymphoma (cHL) is characterized by a rich immune microenvironment as the main tumor component. It involves a broad range of cell populations, which are largely unexplored, even though they are known to be essential for growth and survival of Hodgkin and Reed-Sternberg cells. We profiled the gene expression of 25 FFPE cHL samples using NanoString technology and resolved their microenvironment compositions using cell-deconvolution tools, thereby generating patient-specific signatures. The results confirm individual immune fingerprints and recognize multiple clusters enriched in refractory patients, highlighting the relevance of: (1) the composition of immune cells and their functional status, including myeloid cell populations (M1-like, M2-like, plasmacytoid dendritic cells, myeloid-derived suppressor cells, etc.), CD4-positive T cells (exhausted, regulatory, Th17, etc.), cytotoxic CD8 T and natural killer cells; (2) the balance between inflammatory signatures (such as IL6, TNF, IFN-γ/TGF-β) and MHC-I/MHC-II molecules; and (3) several cells, pathways and genes related to the stroma and extracellular matrix remodeling. A validation model combining relevant immune and stromal signatures identifies patients with unfavorable outcomes, producing the same results in an independent cHL series. Our results reveal the heterogeneity of immune responses among patients, confirm previous findings, and identify new functional phenotypes of prognostic and predictive utility.
Collapse
Affiliation(s)
- Victoria Menéndez
- Translational Research, Fundación MD Anderson International España. Madrid, 28033, Madrid, Spain
| | - José L Solórzano
- Translational Research, Fundación MD Anderson International España. Madrid, 28033, Madrid, Spain
- Pathology Department, MD Anderson Cancer Center Madrid, C/Arturo Soria, 270, 28033, Madrid, Spain
| | - Mónica García-Cosío
- Pathology Department, Hospital Universitario Ramón y Cajal, 28034, Madrid, Spain
| | - Ruth Alonso-Alonso
- Pathology Department, IIS Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), ISCIII, 28029, Madrid, Spain
| | - Marta Rodríguez
- Pathology Department, IIS Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), ISCIII, 28029, Madrid, Spain
| | - Laura Cereceda
- Translational Research, Fundación MD Anderson International España. Madrid, 28033, Madrid, Spain
- Pathology Department, MD Anderson Cancer Center Madrid, C/Arturo Soria, 270, 28033, Madrid, Spain
| | - Sara Fernández
- Translational Research, Fundación MD Anderson International España. Madrid, 28033, Madrid, Spain
- Pathology Department, MD Anderson Cancer Center Madrid, C/Arturo Soria, 270, 28033, Madrid, Spain
| | - Eva Díaz
- Translational Research, Fundación MD Anderson International España. Madrid, 28033, Madrid, Spain
| | - Carlos Montalbán
- Hematology Department, MD Anderson Cancer Center Madrid, 28033, Madrid, Spain
| | - Mónica Estévez
- Hematology Department, MD Anderson Cancer Center Madrid, 28033, Madrid, Spain
| | - Miguel A Piris
- Pathology Department, IIS Hospital Universitario Fundación Jiménez Díaz, 28040, Madrid, Spain
- Center for Biomedical Network Research on Cancer (CIBERONC), ISCIII, 28029, Madrid, Spain
| | - Juan F García
- Translational Research, Fundación MD Anderson International España. Madrid, 28033, Madrid, Spain.
- Pathology Department, MD Anderson Cancer Center Madrid, C/Arturo Soria, 270, 28033, Madrid, Spain.
- Center for Biomedical Network Research on Cancer (CIBERONC), ISCIII, 28029, Madrid, Spain.
| |
Collapse
|
4
|
Georgoulis V, Papoudou-Bai A, Makis A, Kanavaros P, Hatzimichael E. Unraveling the Immune Microenvironment in Classic Hodgkin Lymphoma: Prognostic and Therapeutic Implications. BIOLOGY 2023; 12:862. [PMID: 37372147 PMCID: PMC10294989 DOI: 10.3390/biology12060862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
Classic Hodgkin lymphoma (cHL) is a lymphoid neoplasm composed of rare neoplastic Hodgkin and Reed-Sternberg (HRS) cells surrounded by a reactive tumor microenvironment (TME) with suppressive properties against anti-tumor immunity. TME is mainly composed of T cells (CD4 helper, CD8 cytotoxic and regulatory) and tumor-associated macrophages (TAMs), but the impact of these cells on the natural course of the disease is not absolutely understood. TME contributes to the immune evasion of neoplastic HRS cells through the production of various cytokines and/or the aberrant expression of immune checkpoint molecules in ways that have not been fully understood yet. Herein, we present a comprehensive review of findings regarding the cellular components and the molecular features of the immune TME in cHL, its correlation with treatment response and prognosis, as well as the potential targeting of the TME with novel therapies. Among all cells, macrophages appear to be a most appealing target for immunomodulatory therapies, based on their functional plasticity and antitumor potency.
Collapse
Affiliation(s)
- Vasileios Georgoulis
- Department of Hematology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| | - Alexandra Papoudou-Bai
- Department of Pathology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| | - Alexandros Makis
- Department of Child Health, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 000 Ioannina, Greece;
| | - Eleftheria Hatzimichael
- Department of Hematology, School of Health Sciences, Faculty of Medicine, University of Ioannina, 45 500 Ioannina, Greece;
| |
Collapse
|
5
|
Casagrande N, Borghese C, Aldinucci D. Current and Emerging Approaches to Study Microenvironmental Interactions and Drug Activity in Classical Hodgkin Lymphoma. Cancers (Basel) 2022; 14:cancers14102427. [PMID: 35626032 PMCID: PMC9139207 DOI: 10.3390/cancers14102427] [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: 04/15/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary In classical Hodgkin Lymphoma (cHL), the tumor microenvironment (TME) plays an important role in tumor progression and treatment response, making its evaluation critical for determining prognosis, treatment strategies and predicting an increase in drug toxicity. Therefore, there is a need to utilize more complex systems to study the cHL-TME and its interplay with tumor cells. To evaluate new anticancer drugs and to find the mechanisms of drug resistance, this review summarizes emerging approaches for the analysis of the TME composition and to identify the state of the disease; the in vitro techniques used to determine the mechanisms involved in the building of an immunosuppressive and protective TME; new 3-dimensional (3D) models, the heterospheroids (HS), developed to mimic TME interactions. Here, we describe the present and likely future clinical applications indicated by the results of these studies and propose a classification for the in vitro culture methods used to study TME interactions in cHL. Abstract Classic Hodgkin lymphoma is characterized by a few tumor cells surrounded by a protective and immunosuppressive tumor microenvironment (TME) composed by a wide variety of noncancerous cells that are an active part of the disease. Therefore, new techniques to study the cHL-TME and new therapeutic strategies targeting specifically tumor cells, reactivating the antitumor immunity, counteracting the protective effects of the TME, were developed. Here, we describe new methods used to study the cell composition, the phenotype, and the spatial distribution of Hodgkin and Reed–Sternberg (HRS) cells and of noncancerous cells in tumor tissues. Moreover, we propose a classification, with increasing complexity, of the in vitro functional studies used to clarify the interactions leading not only to HRS cell survival, growth and drug resistance, but also to the immunosuppressive tumor education of monocytes, T lymphocytes and fibroblasts. This classification also includes new 3-dimensional (3D) models, obtained by cultivating HRS cells in extracellular matrix scaffolds or in sponge scaffolds, under non-adherent conditions with noncancerous cells to form heterospheroids (HS), implanted in developing chick eggs (ovo model). We report results obtained with these approaches and their applications in clinical setting.
Collapse
|
6
|
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: 10] [Impact Index Per Article: 3.3] [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
|
7
|
The Hodgkin Lymphoma Immune Microenvironment: Turning Bad News into Good. Cancers (Basel) 2022; 14:cancers14051360. [PMID: 35267668 PMCID: PMC8909875 DOI: 10.3390/cancers14051360] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/09/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
The classic Hodgkin lymphoma (cHL) tumor microenvironment (TME) is by far the most abundant component of tumors and is responsible for most of their biological and clinical characteristics. Recent advances in our knowledge of these networks in cellular interactions allow us to understand that the neoplastic Hodgkin and Reed Sternberg (HRS) cells, although they are in the minority, are the main architects of this dysregulated immune milieu. Here, we review the major changes that have happened in recent years: from TME as a helpless bystander, reflecting an ineffective immune response, to a dynamic tumor-promoting and immunosuppressive element. The HRS cells promote survival through interconnected intrinsic and extrinsic alterations, boosting pro-tumoral signaling pathways through genetic aberrations and autocrine growth signals, in parallel with abnormal cytokine secretion for the recruitment and selection of the best cell partners for this immunosuppressive TME. In turn, cHL is already proving to be the perfect model with which to address an immune checkpoint blockade. Preliminary data demonstrate the utility of druggable key signaling pathways in this ensemble, such as JAK-STAT, NF-κB, and others. In addition, myriad biomarkers predicting a response await validation by new in situ multiplex analytical methods, single-cell gene expression, and other techniques. Together, these components will define the functional phenotypes with which we will elucidate the molecular pathogenesis of the disease and improve the survival of patients who are refractory to conventional therapies.
Collapse
|
8
|
Macrophage Infiltration Correlates with Genomic Instability in Classic Hodgkin Lymphoma. Biomedicines 2022; 10:biomedicines10030579. [PMID: 35327381 PMCID: PMC8945507 DOI: 10.3390/biomedicines10030579] [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: 02/11/2022] [Revised: 02/26/2022] [Accepted: 02/27/2022] [Indexed: 12/10/2022] Open
Abstract
Hodgkin lymphoma (HL) is a biologically diverse group of lymphoid tumors, which accounts for 1% of all de novo neoplasms in the world’s population. It is divided into two main groups: the more common classic Hodgkin lymphoma (cHL) and the less common nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL). cHL is further divided into four subtypes, which differ in morphology and the contents of tumor microenvironment. Macrophages are one of the components of tumor microenvironment known to contribute to creating an immunosuppressive microenvironment, which inhibits the activity of cells expressing granzyme B against tumor cells, even when tumor cells are infected with Epstein–Barr virus (EBV). Our research aimed to explore the association between the specific contents of tumor microenvironment and the genetic anomalies in tumor cells. The presence and the relative percentage of cytotoxic T lymphocytes and macrophages was detected by immunohistochemical staining of the antigens specific for certain cell populations. Fluorescent in situ hybridization was used to detect anomalies in the genome of tumor cells and in situ hybridization was used to detect the presence of EBV. Our results show an association between the number of CD163+ macrophages and the number of TP53 copies or BCL6 gene translocation. Patients who had a higher number of CD163+ macrophages infiltrating tumor tissue and three or higher number of copies of TP53 showed poorer survival. We conclude that the presence of macrophages may contribute to genetic instability in cHL, which drives the progression of cHL and decreases survival of the patients.
Collapse
|
9
|
Abstract
Although CAR T-cell therapy is US Food and Drug Administration-approved for B-cell non-Hodgkin lymphomas, the development of adoptive immunotherapy for the treatment of classic Hodgkin lymphoma (cHL) has not accelerated at a similar pace. Adoptive T-cell therapy with Epstein-Barr virus-specific cytotoxic T lymphocytes and CD30 CAR T cells have demonstrated significant clinical responses in early clinical trials of patients with cHL. Additionally, CD19 and CD123 CAR T cells that target the immunosuppressive tumor microenvironment in cHL have also been investigated. Here we discuss the landscape of clinical trials of adoptive immunotherapy for patients with cHL with a view toward current challenges and novel strategies to improve the development of CAR T-cell therapy for cHL.
Collapse
|
10
|
Zhou Y, Wang S, Tao Y, Chen H, Qin Y, He X, Zhou S, Liu P, Yang J, Yang S, Gui L, Lou N, Zhang Z, Yao J, Han X, Shi Y. Low CCL19 expression is associated with adverse clinical outcomes for follicular lymphoma patients treated with chemoimmunotherapy. J Transl Med 2021; 19:399. [PMID: 34544443 PMCID: PMC8454033 DOI: 10.1186/s12967-021-03078-9] [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: 08/07/2021] [Accepted: 09/11/2021] [Indexed: 11/10/2022] Open
Abstract
Background This study aimed to recognize the hub genes associated with prognosis in follicular lymphoma (FL) treated with first-line rituximab combined with chemotherapy. Method RNA sequencing data of dataset GSE65135 (n = 24) were included in differentially expressed genes (DEGs) analysis. Weighted gene co-expression network analysis (WGCNA) was applied for exploring the coexpression network and identifying hub genes. Validation of hub genes expression and prognosis were applied in dataset GSE119214 (n = 137) and independent patient cohort from Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (n = 32), respectively, by analyzing RNAseq expression data and serum protein concentration quantified by ELISA. The Gene Set Enrichment Analysis (GSEA), gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichments analysis were performed. CIBERSORT was applied for tumor-infiltrating immune cells (TIICs) subset analysis. Results A total of 3260 DEGs were obtained, with 1861 genes upregulated and 1399 genes downregulated. Using WGCNA, eight hub genes, PLA2G2D, MMP9, PTGDS, CCL19, NFIB, YAP1, RGL1, and TIMP3 were identified. Kaplan–Meier analysis and multivariate COX regression analysis indicated that CCL19 independently associated with overall survival (OS) for FL patients treated with rituximab and chemotherapy (HR = 0.47, 95% CI [0.25–0.86], p = 0.014). Higher serum CCL19 concentration was associated with longer progression-free survival (PFS, p = 0.014) and OS (p = 0.039). TIICs subset analysis showed that CCL19 expression had a positive correlation with monocytes and macrophages M1, and a negative correlation with naïve B cells and plasma cells. Conclusion CCL19 expression was associated with survival outcomes and might be a potential prognostic biomarker for FL treated with first-line chemoimmunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03078-9.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Shasha Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yunxia Tao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Haizhu Chen
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yan Qin
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiaohui He
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Shengyu Zhou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Peng Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jianliang Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Sheng Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lin Gui
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Ning Lou
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Zhishang Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 41 Damucang Hutong, Xicheng District, Beijing, 100032, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
11
|
Casagrande N, Borghese C, Favero A, Vicenzetto C, Aldinucci D. Trabectedin overcomes doxorubicin-resistance, counteracts tumor-immunosuppressive reprogramming of monocytes and decreases xenograft growth in Hodgkin lymphoma. Cancer Lett 2020; 500:182-193. [PMID: 33326840 DOI: 10.1016/j.canlet.2020.12.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/25/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022]
Abstract
Classical Hodgkin lymphoma (cHL) tumor cells are surrounded by a protective tumor microenvironment (TME). Trabectedin, an anticancer drug targeting both tumor cells and TME, demonstrated a potent antitumor activity against Hodgkin Reed Sternberg (HRS) cells. It was cytotoxic against cHL cell lines, including the doxorubicin-resistant clones, with subnanomolar IC50 values, and inhibited clonogenic growth and heterospheroid cell viability. It induced necroptosis, caused DNA damage, G2/M cell cycle arrest, and increased reactive oxygen species production. It reduced HRS cell secretion of CCL5, M-CSF, IL-6, IL-13 and TARC, and inhibited migration. Conditioned medium from trabectedin-treated HRS cells was less chemoattractive toward monocytes, mesenchymal stromal cells and lymphocytes, and less effective in educating monocytes to become immunosuppressive macrophages. These monocytes expressed lower levels of indoleamine 2,3-dioxygenase-1, CD206 and PD-L1, secreted lower amounts of IL-10, TARC, and TGF-β, and were less able to inhibit the growth of activated lymphocytes. In vivo, trabectedin inhibited by >75% the growth of cHL murine xenografts with minimal weight loss; tumors of trabectedin-treated mice had fewer TAMs and less angiogenesis. Altogether, this study offers a preclinical rationale for trabectedin use as a new drug candidate in relapsed/refractory cHL patients.
Collapse
Affiliation(s)
- Naike Casagrande
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy
| | - Cinzia Borghese
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy
| | - Andrea Favero
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy
| | - Cristina Vicenzetto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy
| | - Donatella Aldinucci
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, PN, Italy.
| |
Collapse
|
12
|
Werner L, Dreyer JH, Hartmann D, Barros MHM, Büttner-Herold M, Grittner U, Niedobitek G. Tumor-associated macrophages in classical Hodgkin lymphoma: hormetic relationship to outcome. Sci Rep 2020; 10:9410. [PMID: 32523087 PMCID: PMC7287068 DOI: 10.1038/s41598-020-66010-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/11/2020] [Indexed: 12/17/2022] Open
Abstract
Commonly attributed to the prevalence of M2 macrophages, tumor-associated macrophages (TAM) are linked to poor outcome in Hodgkin lymphoma (HL). MYC is supposed to control the expression of M2-specific genes in macrophages, and deficiency in MYC-positive macrophages inhibits tumor growth in mouse models. To verify this hypothesis for HL, seventy-six samples were subjected to immunohistochemical double staining using CD68 or CD163 macrophage-specific antibodies and a reagent detecting MYC. For each cell population, labelled cells were grouped according to low, intermediate and high numbers and related to disease-free survival (DFS) and overall survival (OS). MYC+ cells accounted for 21% and 18% of CD68+ and CD163+ cells, respectively. Numbers of MYC- macrophages were significantly higher in EBV+ cases while no differences were observed for MYC+ macrophages between EBV+ and EBV- cases. Cases with highest numbers of macrophages usually showed worst DFS and OS. In most scenarios, intermediate numbers of macrophages were associated with better outcome than very low or very high numbers. Our observations are reminiscent of the "hormesis hypothesis" and suggest that a relative lack of TAM may allow HL growth while macrophages display an inhibitory effect with increasing numbers. Above a certain threshold, TAM may again support tumor growth.
Collapse
Affiliation(s)
- Laura Werner
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany.
| | | | - David Hartmann
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany
| | | | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen Nuremberg, Erlangen, Germany
| | - Ulrike Grittner
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Gerald Niedobitek
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany.,Institute for Pathology, Sana Klinikum Lichtenberg, Berlin, Germany
| |
Collapse
|
13
|
Roussel M, Lhomme F, Roe CE, Bartkowiak T, Gravelle P, Laurent C, Fest T, Irish JM. Mass cytometry defines distinct immune profile in germinal center B-cell lymphomas. Cancer Immunol Immunother 2020; 69:407-420. [PMID: 31919622 PMCID: PMC7764565 DOI: 10.1007/s00262-019-02464-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/26/2019] [Indexed: 02/08/2023]
Abstract
Tumor-associated macrophage and T-cell subsets are implicated in the pathogenesis of diffuse large B-cell lymphoma, follicular lymphoma, and classical Hodgkin lymphoma. Macrophages provide essential mechanisms of tumor immune evasion through checkpoint ligand expression and secretion of suppressive cytokines. However, normal and tumor-associated macrophage phenotypes are less well characterized than those of tumor-infiltrating T-cell subsets, and it would be especially valuable to know whether the polarization state of macrophages differs across lymphoma tumor microenvironments. Here, an established mass cytometry panel designed to characterize myeloid-derived suppressor cells and known macrophage maturation and polarization states was applied to characterize B-lymphoma tumors and non-malignant human tissue. High-dimensional single-cell analyses were performed using dimensionality reduction and clustering tools. Phenotypically distinct intra-tumor macrophage subsets were identified based on abnormal marker expression profiles that were associated with lymphoma tumor types. While it had been proposed that measurement of CD163 and CD68 might be sufficient to reveal macrophage subsets in tumors, results here indicated that S100A9, CCR2, CD36, Slan, and CD32 should also be measured to effectively characterize lymphoma-specific tumor macrophages. Additionally, the presence of phenotypically distinct, abnormal macrophage populations was closely linked to the phenotype of intra-tumor T-cell populations, including PD-1 expressing T cells. These results further support the close links between macrophage polarization and T-cell functional state, as well as the rationale for targeting tumor-associated macrophages in cancer immunotherapies.
Collapse
Affiliation(s)
- Mikael Roussel
- Laboratoire Hématologie, CHU Pontchaillou, Centre Hospitalier Universitaire de Rennes, Pôle Biologie, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France.
| | - Faustine Lhomme
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France
| | - Caroline E Roe
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Todd Bartkowiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Pauline Gravelle
- Service Anatomie et Cytologie Pathologiques and UMR1037, Toulouse, France
| | - Camille Laurent
- Service Anatomie et Cytologie Pathologiques and UMR1037, Toulouse, France
| | - Thierry Fest
- Laboratoire Hématologie, CHU Pontchaillou, Centre Hospitalier Universitaire de Rennes, Pôle Biologie, 2 rue Henri Le Guilloux, 35033, Rennes, France
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
14
|
Arlt A, von Bonin F, Rehberg T, Perez-Rubio P, Engelmann JC, Limm K, Reinke S, Dullin C, Sun X, Specht R, Maulhardt M, Linke F, Bunt G, Klapper W, Vockerodt M, Wilting J, Pukrop T, Dettmer K, Gronwald W, Oefner PJ, Spang R, Kube D. High CD206 levels in Hodgkin lymphoma-educated macrophages are linked to matrix-remodeling and lymphoma dissemination. Mol Oncol 2020; 14:571-589. [PMID: 31825135 PMCID: PMC7053241 DOI: 10.1002/1878-0261.12616] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/08/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages (Mφ) are abundantly present in the tumor microenvironment and may predict outcome in solid tumors and defined lymphoma subtypes. Mφ heterogeneity, the mechanisms of their recruitment, and their differentiation into lymphoma‐promoting, alternatively activated M2‐like phenotypes are still not fully understood. Therefore, further functional studies are required to understand biological mechanisms associated with human tumor‐associated Mφ (TAM). Here, we show that the global mRNA expression and protein abundance of human Mφ differentiated in Hodgkin lymphoma (HL)‐conditioned medium (CM) differ from those of Mφ educated by conditioned media from diffuse large B‐cell lymphoma (DLBCL) cells or, classically, by macrophage colony‐stimulating factor (M‐CSF). Conditioned media from HL cells support TAM differentiation through upregulation of surface antigens such as CD40, CD163, CD206, and PD‐L1. In particular, RNA and cell surface protein expression of mannose receptor 1 (MRC1)/CD206 significantly exceed the levels induced by classical M‐CSF stimulation in M2‐like Mφ; this is regulated by interleukin 13 to a large extent. Functionally, high CD206 enhances mannose‐dependent endocytosis and uptake of type I collagen. Together with high matrix metalloprotease9 secretion, HL‐TAMs appear to be active modulators of the tumor matrix. Preclinical in ovo models show that co‐cultures of HL cells with monocytes or Mφ support dissemination of lymphoma cells via lymphatic vessels, while tumor size and vessel destruction are decreased in comparison with lymphoma‐only tumors. Immunohistology of human HL tissues reveals a fraction of cases feature large numbers of CD206‐positive cells, with high MRC1 expression being characteristic of HL‐stage IV. In summary, the lymphoma‐TAM interaction contributes to matrix‐remodeling and lymphoma cell dissemination.
Collapse
Affiliation(s)
- Annekatrin Arlt
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany.,Network BMBF eMed MMML-Demonstrators, Regensburg, Germany
| | - Frederike von Bonin
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Thorsten Rehberg
- Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Paula Perez-Rubio
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Julia C Engelmann
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Katharina Limm
- Institute of Functional Genomics, University of Regensburg, Germany
| | - Sarah Reinke
- Department of Pathology, Hematopathology Section, UKSH Campus Kiel, Germany
| | - Christian Dullin
- Institute of Diagnostic and Interventional Radiology, University Medical Centre Göttingen, Germany
| | - Xueni Sun
- Institute of Functional Genomics, University of Regensburg, Germany
| | - Rieke Specht
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Markus Maulhardt
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Franziska Linke
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany
| | - Gertrude Bunt
- Clinical Optical Microscopy, Institute of Neuropathology, University Medical Centre Göttingen, Germany
| | - Wolfram Klapper
- Department of Pathology, Hematopathology Section, UKSH Campus Kiel, Germany
| | - Martina Vockerodt
- Institute of Anatomy and Cell Biology, University Medical Centre Göttingen, Germany
| | - Jörg Wilting
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Institute of Anatomy and Cell Biology, University Medical Centre Göttingen, Germany
| | - Tobias Pukrop
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Germany
| | - Katja Dettmer
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Institute of Functional Genomics, University of Regensburg, Germany
| | - Wolfram Gronwald
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Institute of Functional Genomics, University of Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, Germany
| | - Rainer Spang
- Network BMBF eMed MMML-Demonstrators, Regensburg, Germany.,Statistical Bioinformatics, Institute of Functional Genomics, University of Regensburg, Germany
| | - Dieter Kube
- Clinic of Hematology and Medical Oncology, University Medical Centre Göttingen, Germany.,Network BMBF eMed MMML-Demonstrators, Regensburg, Germany
| |
Collapse
|
15
|
Immune and Inflammatory Cells of the Tumor Microenvironment Represent Novel Therapeutic Targets in Classical Hodgkin Lymphoma. Int J Mol Sci 2019; 20:ijms20215503. [PMID: 31694167 PMCID: PMC6862619 DOI: 10.3390/ijms20215503] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Classical Hodgkin Lymphoma (cHL) is a B-cell malignancy that, typically, responds well to standard therapies. However, patients who relapse after standard regimens or are refractory to induction therapy have a dismal outcome. The implementation of novel therapies such as the anti-CD30 monoclonal antibody Brentuximab Vedotin and immune checkpoint inhibitors has provided curative options for many of these patients. Nonetheless, responses are rarely durable, emphasizing the need for new agents. cHL is characterized by a unique microenvironment in which cellular and humoral components interact to promote tumor survival and dissemination. Knowledge of the complex composition of cHL microenvironment is constantly evolving; in particular, there is growing interest in certain cell subsets such as tumor-associated macrophages, myeloid-derived suppressor cells and neutrophils, all of which have a relevant role in the pathogenesis of the disease. The unique biology of the cHL microenvironment has provided opportunities to develop new drugs, many of which are currently being tested in preclinical and clinical settings. In this review, we will summarize novel insights in the crosstalk between tumor cells and non-malignant inflammatory cells. In addition, we will discuss the relevance of tumor-microenvironment interactions as potential therapeutic targets.
Collapse
|
16
|
Menter T, Tzankov A. Lymphomas and Their Microenvironment: A Multifaceted Relationship. Pathobiology 2019; 86:225-236. [PMID: 31574515 DOI: 10.1159/000502912] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/24/2019] [Indexed: 11/19/2022] Open
Abstract
It has become evident that the microenvironment - lymphocytes, macrophages, fibroblasts as well as the extracellular matrix, cytokines, chemokines, and a plethora of other cells, structures and substances residing in the vicinity of tumor cells - plays an important part in the maintenance of cancer growth and survival. This is also relevant in lymphomas. In this review, we give an outline on the importance of the microenvironment for tumors in general and lymphomas in particular, by highlighting certain basic principles of tumor-microenvironment interaction. The relationship of lymphomas and their microenvironment is multifaceted: lymphoma cells need growth factors and cytokines derived from microenvironmental cells for their sustenance and growth. On the contrary, many lymphomas silence or at least deregulate the immune system to escape recognition and subsequent elimination by immune cells, while giving advantage to suppressive microenvironmental compounds such as M2 polarized macrophages, regulatory T-cells, mast cells, and immunosuppressive fibroblasts. We also give a detailed insight across different lymphoma types to show the variety of tumor-microenvironment interactions. Due to its tremendous importance, the microenvironment has also become a new target for oncologic therapy. The most important finding concerning lymphomas with a focus on immunomodulatory substances is also, therefore, highlighted.
Collapse
Affiliation(s)
- Thomas Menter
- Institute of Medical Genetics and Pathology, University of Basel Hospital, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University of Basel Hospital, Basel, Switzerland,
| |
Collapse
|
17
|
Jimenez O, Barros MH, De Matteo E, Garcia Lombardi M, Preciado MV, Niedobitek G, Chabay P. M1-like macrophage polarization prevails in young children with classic Hodgkin Lymphoma from Argentina. Sci Rep 2019; 9:12687. [PMID: 31481738 PMCID: PMC6722052 DOI: 10.1038/s41598-019-49015-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 08/08/2019] [Indexed: 12/23/2022] Open
Abstract
The microenvironment in classical Hodgkin lymphoma (cHL) comprises a mixture of different types of cells, which are responsible for lymphoma pathogenesis and progression. Even though microenvironment composition in adult cHL has been largely studied, only few groups studied pediatric cHL, in which both Epstein Barr virus (EBV) infection and age may display a role in their pathogenesis. Furthermore, our group described that EBV is significantly associated with cHL in Argentina in patients under the age of 10 years old. For that reason, our aim was to describe the microenvironment composition in 46 pediatric cHL patients. M1-like polarization status prevailed in the whole series independently of EBV association. On the other hand, in children older than 10 years, a tolerogenic environment illustrated by higher FOXP3 expression was proved, accompanied by a macrophage polarization status towards M2. In contrast, in children younger than 10 years, M1-like was prevalent, along with an increase in cytotoxic GrB+ cells. This study supports the notion that pediatric cHL exhibits a particular tumor microenvironment composition.
Collapse
Affiliation(s)
- O Jimenez
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - M H Barros
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany
| | - E De Matteo
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - M Garcia Lombardi
- Oncology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - M V Preciado
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina
| | - G Niedobitek
- Institute for Pathology, Unfallkrankenhaus Berlin, Berlin, Germany
| | - P Chabay
- Multidisciplinary Institute for Investigation in Pediatric Pathologies (IMIPP), CONICET-GCBA, Molecular Biology Laboratory, Pathology Division, Ricardo Gutiérrez Children's Hospital, Buenos Aires, Argentina.
| |
Collapse
|
18
|
Ferrarini I, Rigo A, Zamò A, Vinante F. Classical Hodgkin lymphoma cells may promote an IL-17-enriched microenvironment. Leuk Lymphoma 2019; 60:3395-3405. [PMID: 31304817 DOI: 10.1080/10428194.2019.1636983] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In classical Hodgkin lymphoma (cHL), the significance of the interplay between Hodgkin and Reed-Sternberg cells (HRS) and reactive T cells remains poorly defined. By immunohistochemistry on bioptic cHL specimens, we found that HRS and surrounding T lymphocytes stained positive for IL-17 in 40% of cases. IL-17 was detectable in a similar proportion of patients' sera and correlated with disease burden. Supernatants of KM-H2 and HDLM-2 cHL cell lines guided preferential chemotaxis of CCR6+ T lymphocytes. Coculture of cHL cell lines with PBMC promoted the enrichment of Th17 lymphocytes and Foxp3+/IL-17+ cells, whereas T regulatory cells slightly decreased. Soluble CD30 downmodulated membrane CD30 expression on T cells and contributed to their polarization shift by stimulating IL-17 production and reducing IFN-γ synthesis. Thus, HRS and a number of reactive CD4+ T cells, attracted by tumor-secreted chemokines, produce an IL-17 tumor-shaped inflammatory milieu in a cHL subset.
Collapse
Affiliation(s)
- Isacco Ferrarini
- Department of Medicine, Section of Hematology, Cancer Research & Cell Biology Laboratory, University of Verona, Verona, Italy
| | - Antonella Rigo
- Department of Medicine, Section of Hematology, Cancer Research & Cell Biology Laboratory, University of Verona, Verona, Italy
| | - Alberto Zamò
- Department of Oncology, University of Turin, Turin, Italy
| | - Fabrizio Vinante
- Department of Medicine, Section of Hematology, Cancer Research & Cell Biology Laboratory, University of Verona, Verona, Italy
| |
Collapse
|
19
|
Aldinucci D, Borghese C, Casagrande N. Formation of the Immunosuppressive Microenvironment of Classic Hodgkin Lymphoma and Therapeutic Approaches to Counter It. Int J Mol Sci 2019; 20:ijms20102416. [PMID: 31096713 PMCID: PMC6566335 DOI: 10.3390/ijms20102416] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022] Open
Abstract
Classic Hodgkin lymphoma (cHL) is characterized by a few tumor cells surrounded by a protective, immunosuppressive tumor microenvironment composed of normal cells that are an active part of the disease. Hodgkin and Reed-Sternberg (HRS) cells evade the immune system through a variety of different mechanisms. They evade antitumor effector T cells and natural killer cells and promote T cell exhaustion. Using cytokines and extracellular vesicles, they recruit normal cells, induce their proliferation and "educate" (i.e. reprogram) them to become immunosuppressive and protumorigenic. Therefore, alternative treatment strategies are being developed to target not only tumor cells but also the tumor microenvironment. Here we summarize current knowledge on the ability of HRS cells to build their microenvironment and to educate normal cells to become immunosuppressive. We also describe therapeutic strategies to counteract formation of the tumor microenvironment and related processes leading to T cell exhaustion and repolarization of immunosuppressive tumor-associated macrophages.
Collapse
Affiliation(s)
- Donatella Aldinucci
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| | - Cinzia Borghese
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| | - Naike Casagrande
- Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano (PN), Italy.
| |
Collapse
|
20
|
Galati D, Zanotta S, Corazzelli G, Bruzzese D, Capobianco G, Morelli E, Arcamone M, De Filippi R, Pinto A. Circulating dendritic cells deficiencies as a new biomarker in classical Hodgkin lymphoma. Br J Haematol 2018; 184:594-604. [DOI: 10.1111/bjh.15676] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/08/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Domenico Galati
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Serena Zanotta
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Gaetano Corazzelli
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Dario Bruzzese
- Department of Public Health Università degli Studi di Napoli Federico II Napoli Italia
| | - Gaetana Capobianco
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Emanuela Morelli
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Manuela Arcamone
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| | - Rosaria De Filippi
- Department of Clinical Medicine and Surgery Università degli Studi di Napoli Federico II Napoli Italia
| | - Antonio Pinto
- Haematology‐Oncology and Stem‐Cell Transplantation Unit Department of Haematology and Innovative Therapies Istituto Nazionale Tumori – IRCCS – Fondazione G. Pascale Napoli Italia
| |
Collapse
|
21
|
Casagrande N, Borghese C, Visser L, Mongiat M, Colombatti A, Aldinucci D. CCR5 antagonism by maraviroc inhibits Hodgkin lymphoma microenvironment interactions and xenograft growth. Haematologica 2018; 104:564-575. [PMID: 30309853 PMCID: PMC6395337 DOI: 10.3324/haematol.2018.196725] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
Classic Hodgkin lymphoma tumor cells express a functional CCR5 receptor, and tumor tissues express high CCL5 levels, suggesting that CCL5-CCR5 signaling is involved in tumor-microenvironment formation and tumor growth. Using the CCR5 antagonist, maraviroc, and a neutralizing anti-CCL5 antibody, we found that CCL5 secreted by classic Hodgkin lymphoma cells recruited mesenchymal stromal cells and monocytes. The “education” of mesenchymal stromal cells by tumor cell-conditioned medium enhanced mesenchymal stromal cells’ proliferation and CCL5 secretion. In turn, educated mesenchymal stromal cell-conditioned medium increased the clonogenic growth of tumor cells and monocyte migration, but these effects were reduced by maraviroc. Monocyte education by tumor cell-conditioned medium induced their growth and reprogrammed them towards immunosuppressive tumor-associated macrophages that expressed IDO and PD-L1 and secreted IL-10, CCL17 and TGF-β. Educated monocyte-conditioned medium slowed the growth of phytohemagglutinin-activated lymphocytes. Maraviroc decreased tumor cell growth and synergized with doxorubicin and brentuximab vedotin. A three-dimensional heterospheroid assay showed that maraviroc counteracted both the formation and viability of heterospheroids generated by co-cultivation of tumor cells with mesenchymal stromal cells and monocytes. In mice bearing tumor cell xenografts, maraviroc reduced tumor growth by more than 50% and inhibited monocyte accumulation, without weight loss. Finally, in classic Hodgkin lymphoma human tumor tissues, CCL5 and CD68 expression correlated positively, and patients with high CCL5 levels had poor prognosis. In conclusion, since the present challenges are to find molecules counteracting the formation of the immunosuppressive tumor microenvironment or new, less toxic drug combinations, the repurposed drug maraviroc may represent a new opportunity for classic Hodgkin lym phoma treatment.
Collapse
Affiliation(s)
- Naike Casagrande
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Cinzia Borghese
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Lydia Visser
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMcG), the Netherlands
| | - Maurizio Mongiat
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Alfonso Colombatti
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| | - Donatella Aldinucci
- Unit of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Italy
| |
Collapse
|
22
|
Ruella M, Klichinsky M, Kenderian SS, Shestova O, Ziober A, Kraft DO, Feldman M, Wasik MA, June CH, Gill S. Overcoming the Immunosuppressive Tumor Microenvironment of Hodgkin Lymphoma Using Chimeric Antigen Receptor T Cells. Cancer Discov 2017; 7:1154-1167. [PMID: 28576927 PMCID: PMC5628114 DOI: 10.1158/2159-8290.cd-16-0850] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 04/19/2017] [Accepted: 05/31/2017] [Indexed: 01/03/2023]
Abstract
Patients with otherwise treatment-resistant Hodgkin lymphoma could benefit from chimeric antigen receptor T-cell (CART) therapy. However, Hodgkin lymphoma lacks CD19 and contains a highly immunosuppressive tumor microenvironment (TME). We hypothesized that in Hodgkin lymphoma, CART should target both malignant cells and the TME. We demonstrated CD123 on both Hodgkin lymphoma cells and TME, including tumor-associated macrophages (TAM). In vitro, Hodgkin lymphoma cells convert macrophages toward immunosuppressive TAMs that inhibit T-cell proliferation. In contrast, anti-CD123 CART recognized and killed TAMs, thus overcoming immunosuppression. Finally, we showed in immunodeficient mouse models that CART123 eradicated Hodgkin lymphoma and established long-term immune memory. A novel platform that targets malignant cells and the microenvironment may be needed to successfully treat malignancies with an immunosuppressive milieu.Significance: Anti-CD123 chimeric antigen receptor T cells target both the malignant cells and TAMs in Hodgkin lymphoma, thereby eliminating an important immunosuppressive component of the tumor microenvironment. Cancer Discov; 7(10); 1154-67. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1047.
Collapse
Affiliation(s)
- Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael Klichinsky
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saad S Kenderian
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Olga Shestova
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy Ziober
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel O Kraft
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mariusz A Wasik
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Saar Gill
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
23
|
Roussel M, Irish JM, Menard C, Lhomme F, Tarte K, Fest T. Regulatory myeloid cells: an underexplored continent in B-cell lymphomas. Cancer Immunol Immunother 2017; 66:1103-1111. [PMID: 28689360 PMCID: PMC11029098 DOI: 10.1007/s00262-017-2036-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/22/2017] [Indexed: 12/14/2022]
Abstract
In lymphomas arising from the germinal center, prognostic factors are linked to the myeloid compartment. In particular, high circulating monocyte or myeloid-derived suppressor cell counts are associated with poor prognosis for patients with high-grade B-cell lymphomas. Macrophages with an M2 phenotype are enriched within lymphoma tumors. However, the M1/M2 nomenclature is now deprecated and the clinical impact of this phenotype remains controversial. Across cancer types, myeloid cells are primarily thought to function as immune suppressors during tumor initiation and maintenance, but the biological mechanisms behind the myeloid signatures are still poorly understood in germinal center B-cell lymphomas. Herein, we describe the role and clinical relevance of myeloid cells in B-cell lymphoma and propose innovative approaches to decipher this complex cellular compartment. Indeed, characterization of this heterogeneous cell ecosystem has been largely accomplished with "low-resolution" approaches like morphological evaluation and immunohistochemistry, where cells are characterized using a few proteins and qualitative metrics. High-resolution, quantitative approaches, such as mass cytometry, are valuable to better understand myeloid cell diversity, functions, and to identify potential targets for novel therapies.
Collapse
Affiliation(s)
- Mikael Roussel
- CHU de Rennes, Pole de Biologie, Rennes, France.
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France.
- Laboratoire d'Hématologie, CHU Pontchaillou, 2 rue Henri Le Guilloux, 35033, Rennes Cedex, France.
| | - Jonathan M Irish
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cedric Menard
- CHU de Rennes, Pole de Biologie, Rennes, France
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | | | - Karin Tarte
- CHU de Rennes, Pole de Biologie, Rennes, France
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| | - Thierry Fest
- CHU de Rennes, Pole de Biologie, Rennes, France
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre le Cancer, Rennes, France
| |
Collapse
|
24
|
Dendritic cells in hematological malignancies. Crit Rev Oncol Hematol 2016; 108:86-96. [DOI: 10.1016/j.critrevonc.2016.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/17/2023] Open
|
25
|
Tumor-Infiltrating Macrophages in Post-Transplant, Relapsed Classical Hodgkin Lymphoma Are Donor-Derived. PLoS One 2016; 11:e0163559. [PMID: 27685855 PMCID: PMC5042490 DOI: 10.1371/journal.pone.0163559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/11/2016] [Indexed: 01/16/2023] Open
Abstract
Tumor-associated inflammatory cells in classical Hodgkin lymphoma (CHL) typically outnumber the neoplastic Hodgkin/Reed-Sternberg (H/RS) cells. The composition of the inflammatory infiltrate, particularly the fraction of macrophages, has been associated with clinical behavior. Emerging work from animal models demonstrates that most tissue macrophages are maintained by a process of self-renewal under physiologic circumstances and certain inflammatory states, but the contribution from circulating monocytes may be increased in some disease states. This raises the question of the source of macrophages involved in human disease, particularly that of CHL. Patients with relapsed CHL following allogeneic bone marrow transplant (BMT) provide a unique opportunity to begin to address this issue. We identified 4 such patients in our archives. Through molecular chimerism and/or XY FISH studies, we demonstrated the DNA content in the post-BMT recurrent CHL was predominantly donor-derived, while the H/RS cells were derived from the patient. Where possible to evaluate, the cellular composition of the inflammatory infiltrate, including the percentage of macrophages, was similar to that of the original tumor. Our findings suggest that the H/RS cells themselves define the inflammatory environment. In addition, our results demonstrate that tumor-associated macrophages in CHL are predominantly derived from circulating monocytes rather than resident tissue macrophages. Given the association between tumor microenvironment and disease progression, a better understanding of macrophage recruitment to CHL may open new strategies for therapeutic intervention.
Collapse
|
26
|
Aldinucci D, Celegato M, Casagrande N. Microenvironmental interactions in classical Hodgkin lymphoma and their role in promoting tumor growth, immune escape and drug resistance. Cancer Lett 2016; 380:243-52. [DOI: 10.1016/j.canlet.2015.10.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 12/22/2022]
|
27
|
Englund A, Molin D, Enblad G, Karlén J, Glimelius I, Ljungman G, Amini R. The role of tumour‐infiltrating eosinophils, mast cells and macrophages in Classical and Nodular Lymphocyte Predominant Hodgkin Lymphoma in children. Eur J Haematol 2016; 97:430-438. [DOI: 10.1111/ejh.12747] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Annika Englund
- Department of Women's and Children′s Health, Paediatric Oncology Uppsala University Uppsala Sweden
| | - Daniel Molin
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology Uppsala University Uppsala Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology Uppsala University Uppsala Sweden
| | - Jonas Karlén
- Department of Women's and Children's Health Karolinska Institutet Stockholm Sweden
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology Uppsala University Uppsala Sweden
| | - Gustaf Ljungman
- Department of Women's and Children′s Health, Paediatric Oncology Uppsala University Uppsala Sweden
| | - Rose‐Marie Amini
- Department of Immunology, Genetics and Pathology Unit of Pathology Uppsala University Uppsala Sweden
| |
Collapse
|
28
|
New developments in the pathology of malignant lymphoma: a review of the literature published from October 2014-December 2014. J Hematop 2015; 8:21-29. [PMID: 25798206 PMCID: PMC4357643 DOI: 10.1007/s12308-015-0240-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|