1
|
Wang L, Guo M, Hou S. Advances in primary large B-cell lymphoma of immune-privileged sites. Front Immunol 2025; 16:1533444. [PMID: 40078990 PMCID: PMC11896999 DOI: 10.3389/fimmu.2025.1533444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Primary large B-cell lymphoma of immune-privileged sites (IP-LBCL) encompasses a spectrum of relatively rare aggressive B-cell lymphomas, such as primary central nervous system lymphoma (PCNSL), primary testicular large B-cell lymphoma (PTL), and primary vitreoretinal large B-cell lymphoma (PVRL). Macroscopically, the development of IPI-LBCL may be associated with the dysfunction of meningeal lymphatic vessels (mLVs) and the perivascular channel system formed by astrocytes. Microscopically, mutation in MYD88 and CD79B genes plays a pivotal role in the pathogenesis of IP-LBCL. Pathological examination remains the cornerstone for establishing a diagnosis of IP-LBCL. Moreover, traditional imaging is now supplemented by a suite of advanced diagnostic methods, including cytological, genetic, immunological, multiple omics, and molecular biological, which collectively enhance the diagnostic accuracy of IP-LBCL. Despite these advancements, the high recurrence rates and attendant high mortality rates pose significant challenges to achieving long-term survival in IP-LBCL patients. However, the emergence of novel therapeutic agents, such as Bruton's tyrosine kinase inhibitors (BTKi), immune checkpoint inhibitors, immunomodulators, and anti-CD19 chimeric antigen receptor T (CAR-T) cell therapy, has offered promising new avenues for the treatment of IP-LBCL, demonstrating remarkable anti-tumor efficacy in recent years. This review delves into the epidemiology, pathogenesis mechanisms, diagnosis approaches, therapeutic strategies, and prognosis factors associated with IP-LBCL. It meticulously examines the parallels and divergences between the National Comprehensive Cancer Network (NCCN) and European Society for Medical Oncology (ESMO) guidelines, enhancing the professional comprehension of the complexities inherent to IP-LBCL.
Collapse
Affiliation(s)
- Liao Wang
- Shanxi Bethune Hospital Cancer Center Lymphoma Department, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meiru Guo
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Shuling Hou
- Shanxi Bethune Hospital Cancer Center Lymphoma Department, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
2
|
Young K, Shi WY, Xu-Monette Z, Jia Y, Tzankov A, Go H, Li L, Ponzoni M, Wang Y, Zhai Q, Perry A, Wang S, Wang X, Chiu A, Xu M, Visco C, Dybkaer K, Withers H, Long M, Yuan A, Miao Y, Li J, Macias E, Shuai W, Wang B, Bhagat G, Zu Y, Pan Z, Choi W, Montes-Moreno S, Chen W, van Krieken JH, Møller M, Zhan F, Parsons B, Zhang S, Hsi E, Sohani A, Abramson J, Ferreri A, Xu B, Li Y. Prognostic gene expression and microRNA profiling signatures and genetic alterations in primary testicular diffuse large B-cell lymphoma. RESEARCH SQUARE 2025:rs.3.rs-5732026. [PMID: 39866884 PMCID: PMC11760253 DOI: 10.21203/rs.3.rs-5732026/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Primary testicular diffuse large B-cell lymphoma (PT-DLBCL) is a rare and aggressive lymphoma with molecular heterogeneity not well characterize. In this study, we performed next-generation sequencing analysis for a large number of DNA and RNA samples from patients with PT-DLBCL. DNA sequencing analysis identified ≥ 3 chromosomes with copy number variations (CNVs) and microsatellite instability as prognostic biomarkers, rather than MYD88 mutations and genetic subtypes. Remarkably, targeted RNA-seq analysis in 195 patients revealed that TP53 mutations with a ≥ 40% variant allele frequency had significantly adverse prognostic impact, and that a 150-gene expression signature subdivided PT-DLBCL into two distinct clusters, termed as testicular lymphoma tumor (TLT) and microenvironment (ME) subtypes. The TLT subtype featured upregulation of genes involved in B-cell receptor signaling, cell cycle, DNA damage and repair, higher frequencies of CNVs and MYD88 mutations, elder ages, larger tumor sizes, and significantly poorer survival. Genomic microRNA profiling analysis identified significantly differentially expressed microRNAs between 113 PT-DLBCL and 180 systemic DLBCL patients, and further subdivided the PT-DLBCL cohort by microRNA signatures. The subcohort with upregulation of 16 microRNAs associated with PT-DLBCL and testicular tissue expression had significantly better survival. This study revealed characteristic genetic, gene expression, and microRNA profiles and heterogeneity in PT-DLBCL.
Collapse
Affiliation(s)
- Ken Young
- Duke University Medical Center and Duke Cancer Institute
| | | | | | | | | | - Heounjeong Go
- Asan Medical Center, University of Ulsan College of Medicine
| | - Ling Li
- First Affiliated Hospital of Zhengzhou University
| | | | | | | | | | - Shi Wang
- National University Hospital, Singapore
| | | | | | | | | | | | | | - Mark Long
- Roswell Park Comprehensive Cancer Center
| | | | - Yi Miao
- First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital
| | - Jianyong Li
- First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Personalized Cancer Medicine
| | | | | | | | | | | | | | | | | | | | | | | | - Fenghuang Zhan
- Myeloma Center, University of Arkansas for Medical Sciences
| | | | | | | | | | | | | | - Bing Xu
- The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University
| | | |
Collapse
|
3
|
Mocikova H, Janikova A, Sykorova A, Prochazka V, Pirnos J, Duras J, Kopeckova K, Steinerova K, Pytlik R, Blahovcova P, Salek D, Kozak T, Bachanova V, Belada D. Outcome of patients with diffuse large B-cell lymphoma and testicular involvement - real world data. Ann Hematol 2025; 104:675-684. [PMID: 39352469 PMCID: PMC11868350 DOI: 10.1007/s00277-024-06025-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: 08/02/2024] [Accepted: 09/22/2024] [Indexed: 02/28/2025]
Abstract
Patients with testicular lymphoma are at an increased risk of central nervous system (CNS) disease. Optimal strategy for CNS relapse prevention is unknown. We analyzed treatment strategies, cumulative incidence of CNS relapse and prognosis in 229 patients with diffuse large B-cell lymphoma (DLBCL) and testicular involvement: 157 primary testicular lymphomas (PTL) in clinical stages IE/IIE and 72 patients in advanced stages (T-DLBCL) IIIE/IV. Treatments for PTL vs. T-DLBCL included: rituximab-based chemotherapy (80.9% vs. 90.3%), orchiectomy (94.3% vs. 65.3%) and contralateral testicular irradiation (59.8% vs. 44.4%). Majority (84.3%) received CNS prophylaxis with similar rates of prophylactic methotrexate (intravenous 19.1% vs. 16.6%, intrathecal 40.8% vs. 40.4%, or both 24.2% vs. 27.8%) between PTL and T-DLBCL (p = 0.89). Median follow-up was 51.8 months. CNS relapses occurred in 14 (6.1%) of 63 relapsing patients. The 5-year cumulative incidence of CNS relapse in PTL was 4.5% and in T-DLBCL 12.1%. Median time to CNS relapse was 21.9 months. In univariate analyses, orchiectomy was the single significant factor associated with lower risk of CNS relapse in PTL (HR = 0.11 [95% CI, 0-0.124], p = 0.001). Rituximab significantly reduced CNS relapse risk in T-DLBCL (HR = 0.1002, p = 0.0005). Median progression-free survival (PFS) and overall survival (OS) following CNS relapse was dismal in T-DLBCL compared to PTL (PFS 1.6 vs. 37.8 months, p = 0.04 and OS 2.3 vs. 37.8 months, p = 0.05). This study confirmed a favorable impact of rituximab in prevention of CNS relapse in T-DLBCL. Methotrexate prophylaxis did not alter CNS relapse risk. Prognosis of CNS relapse is particularly poor in T-DLBCL.
Collapse
Affiliation(s)
- Heidi Mocikova
- Fakultni nemocnice Kralovske Vinohrady, Department of Haematology and Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Andrea Janikova
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, University Hospital Brno, Masaryk University Brno, Brno, Czech Republic
| | - Alice Sykorova
- University Hospital and Faculty of Medicine, 4th Department of Internal Medicine- Hematology, Hradec Kralove, Czech Republic
| | - Vit Prochazka
- Faculty of Medicine and Dentistry, Department of Haemato-Oncology, Palacky University, Olomouc, Czech Republic
| | - Jan Pirnos
- Ceske Budejovice Hospital, Ceske Budejovice, Czech Republic
| | - Juraj Duras
- University Hospital and Faculty of Medicine, Department of Hemato-Oncology, Ostrava, Czech Republic
| | - Katerina Kopeckova
- Department of Oncology of the 2nd Faculty of Medicine of Charles University, Motol University Hospital, Prague, Czech Republic
| | - Katerina Steinerova
- Department of Clinical Hematology, University Hospital, Pilsen, Czech Republic
| | - Robert Pytlik
- Cell Therapy Department, Institute of Haematology and Blood Transfusion, Prague, Czech Republic
| | - Petra Blahovcova
- Datacenter of the Czech Lymphoma Study Group, Prague, Czech Republic
| | - David Salek
- Department of Internal Medicine, Hematology and Oncology, Faculty of Medicine, University Hospital Brno, Masaryk University Brno, Brno, Czech Republic
| | - Tomas Kozak
- Fakultni nemocnice Kralovske Vinohrady, Department of Haematology and Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Bachanova
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - David Belada
- University Hospital and Faculty of Medicine, 4th Department of Internal Medicine- Hematology, Hradec Kralove, Czech Republic
| |
Collapse
|
4
|
Tang P, Wang T, Song F, Zhang Y, Zhao Y, Yarmohammadi H, Donadon M, Chen Z. Integrating T-cell inflammation features for prognosis in hepatocellular carcinoma: a novel predictive model. J Gastrointest Oncol 2024; 15:2613-2629. [PMID: 39816015 PMCID: PMC11732361 DOI: 10.21037/jgo-2024-874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death globally and accounts for 75% to 90% of primary liver cancer cases. The high mortality rate of HCC, coupled with the absence of reliable prognostic biomarkers, makes its treatment and prognosis evaluation challenging. The features of the T cell-inflamed microenvironment include active interferon (IFN)-γ signaling and the presence of cytotoxic effector molecules, antigen presentation, and T-cell activating cytokines. Although these features are closely associated with anticancer immunity, their specific roles in HCC remain unclear. This study aimed to investigate the role and prognostic significance of T-cell inflammation (TCI) in HCC patients, providing new insights for clinical diagnosis and treatment strategies. Methods We integrated single-sample gene set enrichment analysis (ssGSEA) and weighted gene coexpression network analysis (WGCNA) to identify the genes associated with TCI at both the single-cell and bulk-transcriptome levels. The HCC TCI-related score (HTCIRS) was developed and assessed with 10 different machine learning algorithms and their combinations, which was followed by validation of the key gene KLF2 in clinical samples and tissue microarrays (TMAs). Results We identified 65 genes associated with TCI, of which 36 were significantly correlated with overall survival (OS). The HTCIRS demonstrated excellent performance in prognostic prediction, revealing differences in biological functions and immune cell infiltration between different risk groups within the tumor microenvironment (TME). Furthermore, KLF2 was identified to be linked to the prognosis of patients with HCC. Conclusions The TCI-related score proposed in this study serves as an important tool for prognostic prediction and personalized treatment of patients with HCC, with KLF2 emerging as a potential biomarker for predicting the prognosis of patients with HCC.
Collapse
Affiliation(s)
- Pengju Tang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Tianlun Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Fei Song
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yu Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yiming Zhao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Hooman Yarmohammadi
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matteo Donadon
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
- Department of Surgery, University Maggiore Hospital della Carità, Novara, Italy
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
5
|
Asadi M, Zafari V, Sadeghi-Mohammadi S, Shanehbandi D, Mert U, Soleimani Z, Caner A, Zarredar H. The role of tumor microenvironment and self-organization in cancer progression: Key insights for therapeutic development. BIOIMPACTS : BI 2024; 15:30713. [PMID: 40256216 PMCID: PMC12008505 DOI: 10.34172/bi.30713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/08/2024] [Accepted: 11/20/2024] [Indexed: 04/22/2025]
Abstract
Introduction The tumor microenvironment (TME) plays a pivotal role in cancer progression, influencing tumor initiation, growth, invasion, metastasis, and response to therapies. This study explores the dynamic interactions within the TME, particularly focusing on self-organization-a process by which tumor cells and their microenvironment reciprocally shape one another, leading to cancer progression and resistance. Understanding these interactions can reveal new prognostic markers and therapeutic targets within the TME, such as extracellular matrix (ECM) components, immune cells, and cytokine signaling pathways. Methods A comprehensive search method was employed to investigate the current academic literature on TME, particularly focusing on self-organization in the context of cancer progression and resistance across the PubMed, Google Scholar, and Science Direct databases. Results Recent studies suggest that therapies that disrupt TME self-organization could improve patient outcomes by defeating drug resistance and increasing the effectiveness of conventional therapy. Additionally, this research highlights the essential of understanding the biophysical properties of the TME, like cytoskeletal alterations, in the development of more effective malignancy therapy. Conclusion This review indicated that targeting the ECM and immune cells within the TME can improve therapy effectiveness. Also, by focusing on TME self-organization, we can recognize new therapeutic plans to defeat drug resistance.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Venus Zafari
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Sanam Sadeghi-Mohammadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ufuk Mert
- Institute of Health Sciences, Department of Basic Oncology, Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayşe Caner
- Department of Basic Oncology, Ege University, Institute of Health Sciences, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Hattori K, Makishima K, Suma S, Abe Y, Suehara Y, Sakamoto T, Kurita N, Ishii R, Matsuoka R, Matsuda M, Tsurubuchi T, Nishikawa R, Tanaka S, Mukasa A, Narita Y, Ichimura K, Nagane M, Takano S, Mathis BJ, Ishikawa E, Matsubara D, Chiba S, Sakata‐Yanagimoto M. Association between microenvironment-related genes and prognosis of primary central nervous system lymphoma. EJHAEM 2024; 5:1201-1214. [PMID: 39691244 PMCID: PMC11647707 DOI: 10.1002/jha2.1046] [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: 05/31/2024] [Revised: 10/06/2024] [Accepted: 10/14/2024] [Indexed: 12/19/2024]
Abstract
Background Primary central nervous system lymphoma (PCNSL) is a rare lymphoid malignancy. Systemic profiling of the PCNSL tumor microenvironment (TME) was previously conducted through gene expression analysis. We investigated the prognostic impact of TME on survival to establish novel prognostic biomarkers in PCNSL patients. Methods We analyzed expression levels of 770 neuroinflammation-related (NFR) genes via NanoString nCounter technology in tumor samples from 30 PCNSL patients. Genes related to the "recurrence group (RG)" or "non-recurrence group (NRG)" were identified and validated using whole transcriptomic analysis of an independent PCNSL cohort (n = 30). Results Forty-five of 770 NFR genes were highly expressed in the RG (3-year overall survival (OS, 22.2%), compared with the NRG group (3-year OS 66.7%). Signatures related to glial cells were enriched in the RG-associated gene set. Multivariate analysis revealed that high expressions of TUBB4A (p = 0.028, HR: 3.88), S100B (p = 0.046, HR: 3.093), and SLC6A1 (p = 0.034, HR: 3.765) were significantly related to death. Expression levels of these three genes were also significantly associated with poor OS in the validation cohort. Immunohistochemical staining against TUBB4A, S100B, and proteins specific to glial cells (GFAP, OLIG2, and CD68) revealed significantly higher positivity in RG glial cells. Conclusion These data suggest that TME-related genes play a crucial role in the pathogenesis of PCNSL, complementing the well-known involvement of the NF-kB signaling pathway. TME targeting, especially glial cell-specific proteins, may thus open new and complementary avenues of therapy for all stages of PCNSL.
Collapse
Affiliation(s)
- Keiichiro Hattori
- Department of HematologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | | | - Sakurako Suma
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Yoshiaki Abe
- Department of HematologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Yasuhito Suehara
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Tatsuhiro Sakamoto
- Department of HematologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Naoki Kurita
- Department of HematologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Ryota Ishii
- Department of BiostatisticsInstitute of MedicineUniversity of TsukubaTsukubaJapan
| | - Ryota Matsuoka
- Department of PathologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
| | - Masahide Matsuda
- Department of NeurosurgeryInstitute of Clinical MedicineUniversity of TsukubaTsukubaJapan
| | - Takao Tsurubuchi
- Department of NeurosurgeryInstitute of Clinical MedicineUniversity of TsukubaTsukubaJapan
| | - Ryo Nishikawa
- Department of Neuro‐Oncology/NeurosurgerySaitama Medical University International Medical CenterSaitamaJapan
| | - Shota Tanaka
- Department of NeurosurgeryGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Akitake Mukasa
- Department of NeurosurgeryGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro‐OncologyNational Cancer Center HospitalTokyoJapan
| | - Koichi Ichimura
- Department of Brain Disease Translational ResearchJuntendo University Graduate School of MedicineTokyoJapan
| | - Motoo Nagane
- Department of NeurosurgeryKyorin University Faculty of MedicineTokyoJapan
| | - Shingo Takano
- Department of NeurosurgeryInstitute of Clinical MedicineUniversity of TsukubaTsukubaJapan
| | - Bryan J. Mathis
- Department of Cardiovascular SurgeryInstitute of MedicineUniversity of TsukubaTsukubaJapan
| | - Eiichi Ishikawa
- Department of NeurosurgeryInstitute of Clinical MedicineUniversity of TsukubaTsukubaJapan
| | - Daisuke Matsubara
- Department of PathologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
| | - Shigeru Chiba
- Department of HematologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
| | - Mamiko Sakata‐Yanagimoto
- Department of HematologyInstitute of MedicineUniversity of TsukubaTsukubaJapan
- Department of HematologyUniversity of Tsukuba HospitalTsukubaJapan
- Division of Advanced Hemato‐OncologyTransborder Medical Research CenterUniversity of TsukubaTsukubaJapan
| |
Collapse
|
7
|
Ikeda D, Oura M, Uehara A, Tabata R, Narita K, Takeuchi M, Machida Y, Matsue K. Prognostic relevance of tumor-infiltrating CD4 + cells and total metabolic tumor volume-based risk stratification in diffuse large B-cell lymphoma. Haematologica 2024; 109:2822-2832. [PMID: 38572548 PMCID: PMC11367203 DOI: 10.3324/haematol.2024.285038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
In order to elucidate the relationship between pretreatment radiomic parameters and the proportions of various tumor-infiltrating (TI) cells, we retrospectively analyzed the association of total metabolic tumor volume (TMTV) and TI cells on biopsied tumor lesions in 171 patients with newly diagnosed diffuse large B-cell lymphoma (DLBCL). The surface markers of TI cells were analyzed by multicolor flow cytometry using a dissected single-cell suspension. In examining the correlation between TI cells and positron-emission tomography-derived parameters (maximum standardized uptake value [SUVmax], total metabolic tumor volume [TMTV], and total lesion glycolysis), intratumoral cell types minimally influenced the results, except for a weak negative correlation between CD4+ cells and SUVmax (R=-0.16, P=0.045). Even for the lesion fluorodeoxyglucose uptake at the biopsied site, CD19+ cells (indicative of malignant burden) showed only a weak correlation with the highest SUV (R=0.21, P=0.009), whereas CD3+ (R=-0.25, P=0.002) and CD4+ cells (R=-0.29, P<0.001) demonstrated a similarly weak inverse correlation. High TMTV and low TI CD4+ cells were independently associated with poor prognosis and their combination identified the most adverse population (3-year progression-free survival: 32.3%, 95% confidence interval [CI]: 19.4-53.7; 3-year overall survival: 48.4%, 95% CI: 33.6-69.6). Moreover, radiomic parameters incorporating the international prognostic index significantly improved the 3-year survival prediction (area under the curve: 0.76, P<0.05) compared to their standalone use. This study underscores the prognostic impact of TI CD4+ cells on DLBCL and suggests that integration of TMTV and TI cell analysis enhances the accuracy of prognostic prediction.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Male
- Female
- Middle Aged
- Prognosis
- Aged
- Adult
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Tumor Burden
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Aged, 80 and over
- Retrospective Studies
- Positron-Emission Tomography
- Risk Assessment
- Young Adult
- Fluorodeoxyglucose F18
Collapse
Affiliation(s)
- Daisuke Ikeda
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba.
| | - Mitsuaki Oura
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Atsushi Uehara
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Rikako Tabata
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Kentaro Narita
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | - Masami Takeuchi
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| | | | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, Chiba
| |
Collapse
|
8
|
Zhao M, Wang L, Wang X, He J, Yu K, Li D. Non-neoplastic cells as prognostic biomarkers in diffuse large B-cell lymphoma: A system review and meta-analysis. TUMORI JOURNAL 2024; 110:227-240. [PMID: 38183180 DOI: 10.1177/03008916231221636] [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] [Indexed: 01/07/2024]
Abstract
The microenvironment of diffuse large B-cell lymphoma (DLBCL) is composed of various components, including immune cells and immune checkpoints, some of which have been correlated with the prognosis of DLBCL, but their results remain controversial. Therefore, we conducted a systematic review and meta-analysis to investigate the association between the microenvironment and prognosis in DLBCL. We searched PubMed, Web of Science, and EMBASE for relevant articles between 2001 and 2022. Twenty-five studies involving 4495 patients with DLBCL were included in the analysis. This meta-analysis confirmed that high densities of Foxp3+Tregs and PD-1+T cells are good indicators for overall survival (OS) in DLBCL, while high densities of programmed cell death protein ligand1(PD-L1)-positive expression cells and T-cell immunoglobulin-and mucin domain-3-containing molecule 3 (TIM-3)-positive expression tumor-infiltrating cells (TILs) play a contrary role in OS. Additionally, higher numbers of T-cell intracytoplasmic antigen-1(TIA-1)-positive expression T cells imply better OS and progression-free survival (PFS), while high numbers of lymphocyte activation gene(LAG)-positive expression TILs predict bad OS and PFS. Various non-tumoral cells in the microenvironment play important roles in the prognosis of DLBCL.
Collapse
MESH Headings
- Humans
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/mortality
- Prognosis
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Min Zhao
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Pathology, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center of Chongqing Medical University, Chongqing, China
| | - Lixing Wang
- Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Xingyu Wang
- Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Juan He
- Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Kuai Yu
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center of Chongqing Medical University, Chongqing, China
- Department of Pathology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Li
- Department of Pathology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Pathology, Chongqing Medical University, Chongqing, China
- Molecular Medicine Diagnostic and Testing Center of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Arora J, Ayyappan S, Yin C, Smith BJ, Lemke-Miltner CD, Wang Z, Farooq U, Weiner GJ. T-cell help in the tumor microenvironment enhances rituximab-mediated NK-cell ADCC. Blood 2024; 143:1816-1824. [PMID: 38457360 PMCID: PMC11076912 DOI: 10.1182/blood.2023023370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/10/2024] Open
Abstract
ABSTRACT Rituximab (RTX) and other monoclonal antibodies (mAbs) that bind directly to malignant cells are of great clinical value but are not effective for all patients. A major mechanism of action of RTX is antibody-dependent cellular cytotoxicity (ADCC) mediated by natural killer (NK) cells. Prior in vitro studies in our laboratory demonstrated that T cells contribute to maintaining the viability and cytotoxic potential of NK cells activated by anti-CD20-coated target B cells. Here, we conducted studies using a novel mouse model and clinical correlative analysis to assess whether T-cell help contribute to RTX-mediated NK-cell ADCC in the tumor microenvironment (TME) in vivo. A humanized mouse model was developed using Raji lymphoma cells and normal donor peripheral blood mononuclear cells that allows for control of T-cell numbers in the lymphoma TME. In this model, NK-cell viability and CD16 and CD25 expression dropped after RTX in the absence of T cells but increased in the presence of T cells. RTX therapy was more effective when T cells were present and was ineffective when NK cells were depleted. In patients with indolent lymphoma, fine needle aspirates were obtained before and ∼1 week after treatment with a RTX-containing regimen. There was a strong correlation between CD4+ T cells as well as total T cells in the pretherapy TME and an increase in NK-cell CD16 and CD25 expression after RTX. We conclude that T-cell help in the TME enhances RTX-mediated NK-cell viability and ADCC.
Collapse
Affiliation(s)
- Jyoti Arora
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, Iowa City, IA
| | - Sabarish Ayyappan
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - Chaobo Yin
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
| | - Brian J. Smith
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Biostatistics, University of Iowa, Iowa City, IA
| | | | - Zhaoming Wang
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
| | - Umar Farooq
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, University of Iowa, Iowa City, IA
| | - George J. Weiner
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA
- Department of Internal Medicine, University of Iowa, Iowa City, IA
| |
Collapse
|
10
|
Ganapathi KA, Nicolae A, Egan C, Geng H, Xi L, Pack SD, McFadden JR, Raffeld M, Jaffe ES, Pittaluga S. Peripheral T-cell lymphomas expressing CD30 and CD15 expand the spectrum of anaplastic large cell lymphoma, ALK-negative. Br J Haematol 2024; 204:1862-1871. [PMID: 38613165 DOI: 10.1111/bjh.19442] [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: 10/12/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024]
Abstract
Peripheral T-cell lymphomas (PTCL) are morphologically and biologically heterogeneous and a subset expresses CD30, including anaplastic large cell lymphomas (ALCL) and a minority of PTCL, not otherwise specified (PTCL, NOS). ALCL with ALK translocations (ALCL, ALK+) are readily identified by routine diagnostic methods, but differentiating ALCL without ALK translocation (ALCL, ALK-) and PTCL, NOS expressing CD30 (PTCL CD30+) can be challenging. Furthermore, rare PTCL co-express CD30 and CD15 (PTCL CD30+CD15+); some resemble ALCL, ALK- while others resemble classic Hodgkin lymphoma. To explore the relationship between PTCL CD30+CD15+ and ALCL, ALK-, we analysed 19 cases of PTCL with CD30 expression, previously diagnosed as ALCL, ALK- (nine cases) and PTCL CD30+CD15+ (10 cases) for DUSP22/IRF4 rearrangements, coding RNA expression and selected transcriptome analysis using the NanoString nCounter gene expression analysis platform. Unsupervised clustering showed no clear segregation between ALCL, ALK- and PTCL CD30+CD15+. Three cases previously classified as PTCL CD30+CD15+ showed DUSP22/IRF4 rearrangements, favouring a diagnosis of ALCL, ALK-. Our results suggest that cases previously designated PTCL CD30+CD15+, likely fall within the spectrum of ALCL, ALK-; additionally, a subset of ALCL, ALK- with DUSP22/IRF4 rearrangement expresses CD15, consistent with previous reports and expands the immunophenotypic spectrum of this lymphoma subgroup.
Collapse
MESH Headings
- Female
- Humans
- Male
- Anaplastic Lymphoma Kinase/genetics
- Anaplastic Lymphoma Kinase/metabolism
- Dual-Specificity Phosphatases/genetics
- Gene Rearrangement
- Interferon Regulatory Factors/genetics
- Interferon Regulatory Factors/metabolism
- Ki-1 Antigen/metabolism
- Ki-1 Antigen/genetics
- Ki-1 Antigen/analysis
- Lewis X Antigen/analysis
- Lewis X Antigen/metabolism
- Lymphoma, Large-Cell, Anaplastic/genetics
- Lymphoma, Large-Cell, Anaplastic/pathology
- Lymphoma, Large-Cell, Anaplastic/diagnosis
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Lymphoma, T-Cell, Peripheral/diagnosis
- Mitogen-Activated Protein Kinase Phosphatases/genetics
Collapse
Affiliation(s)
- Karthik A Ganapathi
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Alina Nicolae
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Caoimhe Egan
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Huimin Geng
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Liqiang Xi
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Svetlana D Pack
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jason R McFadden
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Raffeld
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine S Jaffe
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
11
|
Asadi M, Zarredar H, Zafari V, Soleimani Z, Saeedi H, Caner A, Shanehbandi D. Immune Features of Tumor Microenvironment: A Genetic Spotlight. Cell Biochem Biophys 2024; 82:107-118. [PMID: 37870699 DOI: 10.1007/s12013-023-01192-7] [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: 06/03/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
A tumor represents a highly intricate tissue entity, characterized by an exceptionally complex microenvironment that starkly contrasts with the typical physiological surroundings of healthy tissues. Within this tumor microenvironment (TME), every component and factor assume paramount importance in the progression of malignancy and exerts a pivotal influence on a patient's clinical outcome. One of the remarkable aspects of the TME is its remarkable heterogeneity, not only across different types of cancers but even within the same histological category of tumors. In-depth research has illuminated the intricate interplay between specific immune cells and molecules and the dynamic characteristics of the TME. Recent investigations have yielded compelling evidence that several mutations harbored by tumor cells possess the capacity to instigate substantial alterations in the TME. These mutations, often acting as drivers of tumorigenesis, can orchestrate a cascade of events that remodel the TME, thereby influencing crucial aspects of cancer behavior, including its invasiveness, immune evasion, and response to therapies. It is within this nuanced context that the present study endeavors to provide a concise yet comprehensive summary of how specific mutations, within the genetic landscape of cancer cells, can instigate profound changes in TME features. By elucidating the intricate relationship between genetic mutations and the TME, this research aims to contribute to a deeper understanding of cancer biology. Ultimately, the knowledge gained from this study holds the potential to inform the development of more targeted and effective treatments, thereby offering new hope to patients grappling with the complexities of cancer.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Venus Zafari
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayse Caner
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey.
- The University of Texas, MD Anderson Cancer Center, Houston, USA.
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
12
|
Yu T, Xu-Monette ZY, Lagoo A, Shuai W, Wang B, Neff J, Carrillo LF, Carlsen ED, Pina-Oviedo S, Young KH. Flow cytometry quantification of tumor-infiltrating lymphocytes to predict the survival of patients with diffuse large B-cell lymphoma. Front Immunol 2024; 15:1335689. [PMID: 38348048 PMCID: PMC10859492 DOI: 10.3389/fimmu.2024.1335689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024] Open
Abstract
Introduction Our previous studies have demonstrated that tumor-infiltrating lymphocytes (TILs), including normal B cells, T cells, and natural killer (NK) cells, in diffuse large B-cell lymphoma (DLBCL) have a significantly favorable impact on the clinical outcomes of patients treated with standard chemoimmunotherapy. In this study, to gain a full overview of the tumor immune microenvironment (TIME), we assembled a flow cytometry cohort of 102 patients diagnosed with DLBCL at the Duke University Medical Center. Methods We collected diagnostic flow cytometry data, including the proportion of T cells, abnormal B cells, normal B cells, plasma cells, NK cells, monocytes, and granulocytes in fresh biopsy tissues at clinical presentation, and analyzed the correlations with patient survival and between different cell populations. Results We found that low T cell percentages in all viable cells and low ratios of T cells to abnormal B cells correlated with significantly poorer survival, whereas higher percentages of normal B cells among total B cells (or high ratios of normal B cells to abnormal B cells) and high percentages of NK cells among all viable cells correlated with significantly better survival in patients with DLBCL. After excluding a small number of patients with low T cell percentages, the normal B cell percentage among all B cells, but not T cell percentage among all cells, continued to show a remarkable prognostic effect. Data showed significant positive correlations between T cells and normal B cells, and between granulocytes and monocytes. Furthermore, we constructed a prognostic model based on clinical and flow cytometry factors, which divided the DLBCL cohort into two equal groups with remarkable differences in patient survival and treatment response. Summary TILs, including normal B cells, T cells, and NK cells, are associated with favorable clinical outcomes in DLBCL, and flow cytometry capable of quantifying the TIME may have additional clinical utility for prognostication.
Collapse
Affiliation(s)
- Tiantian Yu
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Zijun Y. Xu-Monette
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
- Duke University Cancer Institute, Durham, NC, United States
| | - Anand Lagoo
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Wen Shuai
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Bangchen Wang
- Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Jadee Neff
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Luis F. Carrillo
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Eric D. Carlsen
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
- Duke University Cancer Institute, Durham, NC, United States
| | - Sergio Pina-Oviedo
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
| | - Ken H. Young
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, United States
- Duke University Cancer Institute, Durham, NC, United States
| |
Collapse
|
13
|
Leivonen SK, Friman T, Autio M, Vaittinen S, Jensen AW, D'Amore F, Hamilton-Dutoit SJ, Holte H, Beiske K, Kovanen PE, Räty R, Leppä S. Characterization and clinical impact of the tumor microenvironment in post-transplant aggressive B-cell lymphomas. Haematologica 2023; 108:3044-3057. [PMID: 37259566 PMCID: PMC10620595 DOI: 10.3324/haematol.2023.282831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023] Open
Abstract
Post-transplant lymphoproliferative disorders (PTLD) are iatrogenic immune deficiency-associated lymphoid/plasmacytic proliferations developing due to immunosuppression in solid organ or hematopoietic stem cell allograft patients. PTLD are characterized by abnormal proliferation of lymphoid cells and have a heterogeneous clinical behavior. We profiled expression of >700 tumor microenvironment (TME)-related genes in 75 post-transplant aggressive B-cell lymphomas (PTABCL). Epstein-Barr virus (EBV)-positive PT-ABCL clustered together and were enriched for type I interferon pathway and antiviral-response genes. Additionally, a cytotoxicity gene signature associated with EBV-positivity and favorable overall survival (OS) (hazard ratio =0.61; P=0.019). In silico immunophenotyping revealed two subgroups with distinct immune cell compositions. The inflamed subgroup with higher proportions of immune cells had better outcome compared to noninflamed subgroup (median OS >200.0 vs. 15.2 months; P=0.006). In multivariable analysis with EBV status, International Prognostic Index, and rituximab-containing treatment, inflamed TME remained as an independent predictor for favorable outcome. We also compared TME between post-transplant and immunocompetent host diffuse large B-cell lymphomas (n=75) and discovered that the proportions of T cells were lower in PT-diffuse large B-cell lymphomas. In conclusion, we provide a comprehensive phenotypic characterization of PT-ABCL, highlighting the importance of immune cell composition of TME in determining the clinical behavior and prognosis of PT-ABCL.
Collapse
Affiliation(s)
- Suvi-Katri Leivonen
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki
| | - Terhi Friman
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center and University of Helsinki
| | - Matias Autio
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki
| | - Samuli Vaittinen
- Department of Pathology, Turku University Hospital, University of Turku, Turku
| | | | | | | | - Harald Holte
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Klaus Beiske
- Department of Pathology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Panu E Kovanen
- Department of Pathology, University of Helsinki, and HUSLAB, Helsinki University Hospital, Helsinki
| | - Riikka Räty
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center and University of Helsinki
| | - Sirpa Leppä
- Applied Tumor Genomics Research Program, Medical Faculty, University of Helsinki, Helsinki, Finland; Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki.
| |
Collapse
|
14
|
Yang S, Chang W, Zhang B, Shang P. What factors are associated with the prognosis of primary testicular diffuse large B-cell lymphoma? A study based on the SEER database. J Cancer Res Clin Oncol 2023; 149:10269-10278. [PMID: 37270733 DOI: 10.1007/s00432-023-04907-8] [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: 04/20/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023]
Abstract
PURPOSE Primary testicular diffuse large B-cell lymphoma (PT-DLBCL) is a relatively rare urological tumor with a high degree of malignancy and a poor prognosis. This study aimed to investigate the prognostic risk factors for survival of patients with PT-DLBCL, and then to construct a predictive model and verify its reliability. METHODS First, we selected subjects from the SEER database (2000-2018) and analyzed the survival of PT-DLBCL patients by Kaplan-Meier test. Then, we analyzed prognostic factors by Cox regression. Finally, the data from the training cohort were used to construct a prediction model and represented with a nomogram. We evaluated the nomogram using the consistency index (C-index), decision curve analysis (DCA), and the area under the subject operating characteristic curve (ROC). In addition, calibration curves were plotted to assess the agreement between the column plot model and the actual model. RESULTS We identified five independent risk factors for patient prognosis affecting OS and CSS in patients with PT-DLBCL by univariate and multivariate analysis, including age, transversality, Ann Arbor staging, chemotherapy, and radiotherapy. According to the above factors, we constructed prognostic nomograms, and found that age contributed the most to the survival of patients with PT-DLBCL. The C-indexes for the nomogram of OS and CSS in the training cohort were 0.758 (0.716-0.799) and 0.763 (0.714-0.812), and in the validation cohort were OS and CSS 0.756 (0.697-0.815) and 0.748 (0.679-0.817). CONCLUSION We produced the first nomogram of PT-DLBCL, and it can be used to evaluate the CSS and OS of patients to determine the prognosis of patients.
Collapse
Affiliation(s)
- Shujun Yang
- Department of Urology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Wei Chang
- Department of Urology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Bin Zhang
- Department of Urology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Panfeng Shang
- Department of Urology, Lanzhou University Second Hospital, Chengguan District, Lanzhou, 730030, Gansu, China.
| |
Collapse
|
15
|
Perdikis-Prati S, Sheikh S, Bouroumeau A, Lang N. Efficacy of Immune Checkpoint Blockade and Biomarkers of Response in Lymphoma: A Narrative Review. Biomedicines 2023; 11:1720. [PMID: 37371815 DOI: 10.3390/biomedicines11061720] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Immune checkpoint blockade (ICB) has revolutionized the prognosis of several advanced-stage solid tumors. However, its success has been far more limited in hematological malignancies and is mostly restricted to classical Hodgkin lymphoma (cHL) and primary mediastinal B cell lymphoma (PMBCL). In patients with non-Hodgkin lymphoma (NHL), response to PD-1/PD-L1 ICB monotherapy has been relatively limited, although some subtypes are more sensitive than others. Numerous predictive biomarkers have been investigated in solid malignancies, such as PD-L1 expression, tumor mutational burden (TMB) and microsatellite instability (MSI), among others. This review aims to appraise the current knowledge on PD-1/PD-L1 ICB efficacy in lymphoma when used either as monotherapy or combined with other agents, and describes potential biomarkers of response in this specific setting.
Collapse
Affiliation(s)
| | - Semira Sheikh
- Department of Hematology, Universitätsspital Basel, 4031 Basel, Switzerland
| | - Antonin Bouroumeau
- Division of Clinical Pathology, Diagnostic Department, Geneva University Hospital, 1206 Geneva, Switzerland
| | - Noémie Lang
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
- Center of Translational Research in Oncohematology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| |
Collapse
|
16
|
Koviazin AK, Filatova LV, Zyuzgin IS, Artemyeva AS, Poliatskin IL, Burda DS, Volchenkov SA, Elkhova SS, Semiglazova TY. The significance of upfront autologous stem cell transplantation for high-intermediate/high-risk stage IV diffuse large B-cell lymphoma. Cancer Rep (Hoboken) 2023; 6:e1786. [PMID: 36855295 PMCID: PMC10075296 DOI: 10.1002/cnr2.1786] [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: 08/31/2022] [Revised: 12/03/2022] [Accepted: 12/31/2022] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is the most common (30%-35%) type of B-cell lymphoma. Only about 60% of all newly diagnosed advanced-stage DLBCL can be completely treated with x6 R-CHOP. High-dose chemotherapy (HDCT) followed by autologous hematopoietic stem cell transplantation in the first remission (upfront auto-HSCT) can serve as an option to improve a prognosis in these patients. AIMS This trial aimed to improve prognosis in DLBCL by upfront auto-HSCT. METHODS AND RESULTS A group of 105 patients: DLBCL NOS, age 18-65, stage IV, IPI ≥2, CR/PR after x6 R-CHOP/DA-EPOCH-R from 2010 to 2019 at NMRC of Oncology named after N.N.Petrov of MoH of Russia was retrospectively analyzed. The HSCT group included patients with upfront HDCT followed by auto-HSCT (n = 35). The control group included patients with non-invasive follow-up after induction (n = 70). Primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), response rate and relapse rate. The 3-year OS (p = .013) and 3-year PFS (p = .033) were significantly higher in the HSCT group. The 3-year OS was decreased by the occurrence of relapse (p ≤ .001) and weight loss (B-symptom) (p = .04). DEL was the negative prognostic factor for 3-year PFS in all patients (p = .001) and control group (p = .001). DA-EPOCH-R significantly increased the 3-year PFS (p = .041). CONCLUSION Upfront HDCT followed by auto-HSCT can increase 3-year OS and PFS and improve prognosis in DLBCL NOS, age 18-65, stage IV, IPI ≥2 patients.
Collapse
Affiliation(s)
- Aleksei K Koviazin
- Department of Hematology and Chemotherapy with Intensive Care Unit, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation.,Department of Innovative Methods in Therapeutic Oncology and Rehabilitation, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Larisa V Filatova
- Department of Hematology and Chemotherapy with Intensive Care Unit, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation.,Department of Innovative Methods in Therapeutic Oncology and Rehabilitation, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation.,Department of Oncology, Federal State Budgetary Educational Institution of Higher Education "North-Western State Medical University named after I.I. Mechnikov" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Ilia S Zyuzgin
- Department of Hematology and Chemotherapy with Intensive Care Unit, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Anna S Artemyeva
- Laboratory of Tumor Morphology, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Ilia L Poliatskin
- Laboratory of Tumor Morphology, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Darya S Burda
- Laboratory of Tumor Morphology, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Stanislav A Volchenkov
- Department of Hematology and Chemotherapy with Intensive Care Unit, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Svetlana S Elkhova
- Department of Hematology and Chemotherapy with Intensive Care Unit, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| | - Tatiana Yu Semiglazova
- Department of Innovative Methods in Therapeutic Oncology and Rehabilitation, NMRC of Oncology n.a. N.N.Petrov of MoH of Russia, Federal State Budgetary Institution "Petrov National Medical Cancer Research Centre" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation.,Department of Oncology, Federal State Budgetary Educational Institution of Higher Education "North-Western State Medical University named after I.I. Mechnikov" of the Ministry of Health of the Russian Federation, Saint-Petersburg, Russian Federation
| |
Collapse
|
17
|
Li W, Lv L, Ruan M, Xu J, Zhu W, Li Q, Jiang X, Zheng L, Zhu W. Qin Huang formula enhances the effect of Adriamycin in B-cell lymphoma via increasing tumor infiltrating lymphocytes by targeting toll-like receptor signaling pathway. BMC Complement Med Ther 2022; 22:185. [PMID: 35818037 PMCID: PMC9272877 DOI: 10.1186/s12906-022-03660-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 06/29/2022] [Indexed: 12/05/2022] Open
Abstract
Background As an original traditional Chinese medicinal formula, Qin Huang formula (QHF) is used as adjuvant therapy for treating lymphoma in our hospital and has proven efficacy when combined with chemotherapy. However, the underlying mechanisms of QHF have not been elucidated. Methods A network pharmacological-based analysis method was used to screen the active components and predict the potential mechanisms of QHF in treating B cell lymphoma. Then, a murine model was built to verify the antitumor effect of QHF combined with Adriamycin (ADM) in vivo. Finally, IHC, ELISA, 18F-FDG PET-CT scan, and western blot were processed to reveal the intriguing mechanism of QHF in treating B cell lymphoma. Results The systemic pharmacological study revealed that QHF took effect following a multiple-target and multiple-pathway pattern in the human body. In vivo study showed that combination therapy with QHF and ADM potently inhibited the growth of B cell lymphoma in a syngeneic murine model, and significantly increased the proportion of tumor infiltrating CD4+ and CD8+ T cells in the tumor microenvironment (TME). Furthermore, the level of CXCL10 and IL-6 was significantly increased in the combination group. Finally, the western blot exhibited that the level of TLR2 and p38 MAPK increased in the combination therapy group. Conclusion QHF in combination of ADM enhances the antitumor effect of ADM via modulating tumor immune microenvironment and can be a combination therapeutic strategy for B cell lymphoma patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03660-8.
Collapse
|
18
|
Stubbins RJ, Lam R, Zhu J, Ghosh S, Mabilangan C, Kuruvilla J, Goswami RS, Lai R, Preiksaitis JK, Jain MD, Peters AC. Tumor Infiltrating Lymphocytes Predict Survival in Solid Organ Transplant Recipients With Monomorphic Post-transplant Lymphoproliferative Disorders. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:744-752. [PMID: 35717340 DOI: 10.1016/j.clml.2022.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION The tumor microenvironment (TME) in post-transplant lymphoproliferative disorders (PTLDs) remains unexplored. Tumor infiltrating lymphocytes (TILs) are prognostic in other lymphomas. We assessed the prognostic impact of TILs in monomorphic B-cell PTLD. METHODS TIL density (CD3+ cells/mm2) was determined by CD3 immunohistochemistry in archived diagnostic biopsies from patients diagnosed with monomorphic B-cell PTLD. RESULTS Amongst monomorphic PTLDs (N = 107), low TIL-count was associated with inferior 2-year progression-free survival (PFS) (41% versus 86%, P = .003) and 2-year overall survival (OS) (52% versus 93%, P = .003) by Kaplan-Meier analysis. Low TIL-count was significant on Cox univariate regression for inferior PFS (HR 4.5, 95% CI 2.0-9.9, P < .001) and OS (HR 4.6, 95% CI 1.8-11.8, P < .001). Multivariate analysis with clinical variables (age ≥60 years, high LDH, stage III/IV, CNS involvement) and TIL-count showed significance for PFS (HR 3.3, 95% CI 1.3-8.3, P = .010) and a non-significant trend for OS (HR 2.6, 95% CI 0.9-7.3, P = .064). A composite score including TILs and clinical variables (age ≥60 years, high LDH, stage III/IV, CNS involvement) effectively stratified monomorphic PTLD patients by PFS and OS (2-year OS: low-risk 93%, intermediate-risk 61%, high-risk 23%, P < .001). CONCLUSIONS The TME and TILs are prognostically relevant in monomorphic PTLD. Prognostic models including measures of the TME may improve risk stratification for patients with monomorphic PTLDs.
Collapse
Affiliation(s)
- Ryan J Stubbins
- Leukemia/BMT Program of BC, BC Cancer, Vancouver, BC, Canada
| | - Ryan Lam
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - James Zhu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Sunita Ghosh
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Curtis Mabilangan
- Division of Infectious Diseases, Department of Medicine, University of Alberta, AB, Edmonton, Canada
| | - John Kuruvilla
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rashmi S Goswami
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Jutta K Preiksaitis
- Division of Infectious Diseases, Department of Medicine, University of Alberta, AB, Edmonton, Canada
| | - Michael D Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute; Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Anthea C Peters
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
19
|
Jiang Y, Sun H, Xu H, Hu X, Wu W, Lv Y, Wang J, Liu S, Zhai Y, Tian L, Wang Y, Zhao Z. Immunophenotypic Landscape and Prognosis-Related mRNA Signature in Diffuse Large B Cell Lymphoma. Front Genet 2022; 13:872001. [PMID: 35754837 PMCID: PMC9214219 DOI: 10.3389/fgene.2022.872001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/20/2022] [Indexed: 11/25/2022] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) exhibits a tightly complexity immune landscape. In this study, we intended to identify different immune phenotype and to examine the immune related mRNA signature for clinical characteristic, therapeutic responsiveness as well as risk stratification and survival prediction in DLBCL. We identified two immune infiltration subtypes of DLBCL patients based on 28 immune cell types. GSEA analysis uncovered the concordant classification of two robust significant subtypes of DLBCL. Considering the convenient application of the immune infiltration subtypes for prognostic prediction, we developed a risk score based on the differentially expressed genes between the Immunity-H and Immunity-L groups. By a least absolute shrinkage and selection operator (LASSO)-Cox regression model, a sixteen-gene risk signature, comprising ANTXR1, CD3D, TIMP1, FPR3, NID2, CTLA4, LPAR6, GPR183, LYZ, PTGDS, ITK, FBN1, FRMD6, PLAU, MICAL2, C1S, was established. The comprehensive results showed that the high-risk group was correlated with lower immune infiltration, more aggressive phenotypes, lower overall survival and more sensitive to lenalidomide. In contrast, a low-risk group score was associated with higher immune infiltration, less aggressive phenotypes, better overall survival and more likely to benefit from PD-1/PD-L1 inhibitors. Finally, a nomogram comprised of the risk score and IPI score was verified to more accurately predict the overall survival of DLBCL than traditional clinical prediction models. Altogether, our data demonstrate the heterogeneity of immune patterns within DLBCL and deepen our molecular understanding of this tumor entity.
Collapse
Affiliation(s)
- Yanan Jiang
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Huimeng Sun
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hong Xu
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xin Hu
- Key Laboratory of Cancer Prevention and Therapy of Tianjin, Key Laboratory of Molecular Cancer Epidemiology, Department of Epidemiology and Biostatistics, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wenqi Wu
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yangyang Lv
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinhuan Wang
- Department of Oncology, Institute of Urology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Su Liu
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yixin Zhai
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Linyan Tian
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yafei Wang
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhigang Zhao
- Key Laboratory of Cancer Prevention and Therapy, Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
20
|
Li Y, Zhang Y, Wang W, Wei C, Zhao D, Zhang W. Follicular Lymphoma in China: Systematic Evaluation of Follicular Lymphoma Prognostic Models. Cancer Manag Res 2022; 14:1385-1393. [PMID: 35422658 PMCID: PMC9005237 DOI: 10.2147/cmar.s349193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/31/2022] [Indexed: 01/18/2023] Open
Abstract
Objects To systematically evaluate the baseline characteristics, tumour microenvironment indicators such as the lymphocyte/monocyte ratio (LMR) and treatment response (POD24) as prognostic predictors of follicular lymphoma (FL) among Chinese patients. Methods We retrospectively analysed 112 FL patients from 2000 to 2017, whose pathology grading included 1-3a. Absolute lymphocyte and monocyte counts were determined by cell blood counting. The Kaplan-Meier method was used to analyse the influence of prognostic predictors on progression-free survival (PFS) and overall survival (OS). Multivariate analysis was conducted using the Cox proportional risk model. Results The long-term survival of Chinese FL patients (median PFS, 74.8 months) was slightly better than that of patients in foreign countries. Among several prognostic models, the Follicular Lymphoma International Prognostic Index (FLIPI) score had a significant effect on both PFS and OS, while there were no independent prognostic predictors. Patients with LMR ≤3.6, ALC ≤0.6*109/L and AMC >0.6*109/L had worse OS, among which LMR was an independent indicator of OS. The POD24 subgroup had a higher frequency of high-risk patients according to FLIPI and FLIPI2 scores (63.0% vs 35.1%, P = 0.013 and 18.5% vs 3.9%, P = 0.003) and had an markedly shorter OS (P < 0.0001). Conclusion This study is a systematic prognostic evaluation based on Chinese clinical data. We found a new factor, the LMR, that could independently predict prognosis compared with FLIPI or FLIPI2.
Collapse
Affiliation(s)
- Yanan Li
- Department of Hematology, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| | - Yan Zhang
- Department of Hematology, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| | - Wei Wang
- Department of Hematology, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| | - Chong Wei
- Department of Hematology, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| | - Danqing Zhao
- Department of Hematology, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| | - Wei Zhang
- Department of Hematology, Peking Union Medical College Hospital, Beijing, People’s Republic of China
| |
Collapse
|
21
|
Wang Z, Zhong Y, Zhang Z, Zhou K, Huang Z, Yu H, Liu L, Liu S, Yang H, Zhou J, Fan J, Wu L, Sun Y. Characteristics and Clinical Significance of T-Cell Receptor Repertoire in Hepatocellular Carcinoma. Front Immunol 2022; 13:847263. [PMID: 35371059 PMCID: PMC8965762 DOI: 10.3389/fimmu.2022.847263] [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: 01/01/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Several studies have demonstrated that the T-cell receptor (TCR) repertoire is associated with prognosis and immune therapy response in several types of cancer. However, the comprehensive features of TCR repertoire in tumor-infiltrating and circulating T cells, as well as its clinical significance of diagnosis in hepatocellular carcinoma (HCC) patients, are still unknown. In this study, we perform paired tumor/peritumoral tissues and peripheral blood samples from 58 patients with HCC and sequenced them with high-throughput TCR to comprehensively analyze the characteristics of TCR and the clinical significance of peripheral TCR sequence. By exploring the abundance and diversity of TCR repertoires, we observe that there was a significantly higher TCR diversity in peripheral blood than in tumoral and peritumoral tissues, while tumoral and peritumoral tissues showed similar TCR diversity. A substantial difference in the usage frequencies of several Vβ, Jβ genes, and TCRβ VJ pairings was found among three types of tissues. Moreover, we reveal that HCC patients have a unique profile of TCR repertoire in peripheral blood in contrast to healthy individuals. We further establish an HCC diagnostic model based on TCRβ VJ pairing usage in peripheral blood, which yields a best-fit area under the curve (AUC) of 0.9746 ± 0.0481 (sensitivity = 0.9675 ± 0.0603, specificity = 0.9998 ± 0.0007, average of 100 repeats) in the test set. Our study describes the characteristics of tissue infiltration and circulating T-cell bank in patients with HCC and shows the potential of using circulating TCR sequence as a biomarker for the non-invasive diagnosis of patients with HCC.
Collapse
Affiliation(s)
- Zifei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
| | - Yu Zhong
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
| | - Zefan Zhang
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Kaiqian Zhou
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Zhihao Huang
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
| | - Hao Yu
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
| | - Longqi Liu
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| | - Shiping Liu
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Liang Wu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| | - Yunfan Sun
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
22
|
Chen W, Liang W, He Y, Liu C, Chen H, Lv P, Yao Y, Zhou H. Immune microenvironment-related gene mapping predicts immunochemotherapy response and prognosis in diffuse large B-cell lymphoma. Med Oncol 2022; 39:44. [PMID: 35092504 DOI: 10.1007/s12032-021-01642-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/23/2021] [Indexed: 01/01/2023]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin's lymphoma (NHL). The R-CHOP immunochemotherapy regimen is the first-line treatment option for DLBCL patients and has greatly improved the prognosis of DLBCL, making it a curable disease. However, drug resistance or relapse is the main challenge for current DLBCL treatment. Studies have shown that the tumor microenvironment plays an important role in the onset, development, and responsiveness to drugs in DLBCL. Here, we used the CIBERSORT algorithm to resolve the composition of the immune microenvironment of 471 DLBCL patients from the GEO database. We found that activated memory CD4+ T cells and γδ T cells were significantly associated with immunochemotherapy response. Weighted gene co-expression networks (WGCNA) were constructed using differentially expressed genes from immunochemotherapy responders and non-responders. The module most associated with these two types of T cells was defined as hub module. Enrichment analysis of the hub module showed that baseline immune status was significantly stronger in responders than in non-responders. A protein-protein interaction (PPI) network was constructed for hub module to identify hub genes. After survival analysis, five prognosis-related genes (CD3G, CD3D, GNB4, FCHO2, GPR183) were identified and all these genes were significantly negatively associated with PD1. Using our own patient cohort, we validated the efficacy of CD3G and CD3D in predicting immunochemotherapy response. Our study showed that CD3G, CD3D, GNB4, FCHO2, and GPR183 are involved in the regulation of the immune microenvironment of DLBCL. They can be used as biomarkers for predicting immunochemotherapy response and potential therapeutic targets in DLBCL.
Collapse
Affiliation(s)
- Wanjun Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weijie Liang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongjian He
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chixiang Liu
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongtian Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Piao Lv
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Yao
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huayou Zhou
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
23
|
Yan Z, Yao S, Wang Y, Liu Y, Yao Z. Primary Testicular Lymphoma with Central Nervous System Relapse Was Successfully Treated by a Chemo-Free Regimen: A Case Report and Literature Review. Cancer Manag Res 2022; 13:9489-9500. [PMID: 35002326 PMCID: PMC8725687 DOI: 10.2147/cmar.s341342] [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: 10/04/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Primary testicular lymphoma (PTL) is a rare malignancy of testis. Although the multimodality treatment (including orchiectomy, systemic chemotherapy, scrotal radiotherapy, and preventive central nervous system (CNS)-targeted treatment) is widely used to treat PTL, recurrence, especially CNS recurrence, occurred frequently. Patients with relapsed PTL have a dismal prognosis and limited treatment options. In this report, we described the case of a 63-year-old man with early-stage PTL. The patient received the multimodality treatment, but CNS relapse occurred 3 months following the front-line therapy. We gave him a combined chemo-free regimen treatment, including rituximab, ibrutinib, and lenalidomide (RIL), based on the tumor's gene mutation profile and the patient's preference. A complete response was achieved after the first cycle of treatment. Whole-brain radiotherapy was delivered as consolidative treatment following three more cycles of RIL. Thereafter, ibrutinib and lenalidomide continued as maintenance treatment. As of the submission of this manuscript, the response has lasted for more than 16 months. Based on the case, we believe chemo-free regimen RIL might be a favorable approach for PTL patients with CNS relapse, especially those frail elderly patients, when alternative treatments are not available.
Collapse
Affiliation(s)
- Zheng Yan
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shuna Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yuanyuan Wang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yanyan Liu
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhihua Yao
- Department of Internal Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
24
|
Checkpoint protein expression in the tumor microenvironment defines the outcome of classical Hodgkin lymphoma patients. Blood Adv 2021; 6:1919-1931. [PMID: 34941990 PMCID: PMC8941476 DOI: 10.1182/bloodadvances.2021006189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/28/2021] [Indexed: 11/20/2022] Open
Abstract
Emerging evidence indicates a major impact for the tumor microenvironment (TME) and immune escape in the pathogenesis and clinical course of classical Hodgkin lymphoma (cHL). We used gene expression profiling (n=88), CIBERSORT, and multiplex immunohistochemistry (n=131) to characterize the immunoprofile of cHL TME, and correlated the findings with survival. Gene expression analysis divided tumors into subgroups with T cell-inflamed and non-inflamed TME. Several macrophage-related genes were upregulated in samples with the non-T cell-inflamed TME, and based on the immune cell proportions, the samples clustered according to the content of T cells and macrophages. A cluster with high proportions of checkpoint protein (PD-1, PD-L1, IDO-1, LAG-3, and TIM-3) positive immune cells translated to unfavorable overall survival (OS) (5-year OS 76% vs. 96%, P=0.010), and remained as an independent prognostic factor for OS in multivariable analysis (HR 4.34, 95% CI 1.05-17.91, P=0.043). cHLs with high proportions of checkpoint proteins overexpressed genes coding for cytolytic factors, proposing paradoxically that they were immunologically active. This checkpoint molecule gene signature translated to inferior survival in a validation cohort of 290 diagnostic cHL samples (P<0.001) and in an expansion cohort of 84 cHL relapse samples (P=0.048). Our findings demonstrate the impact of T cell- and macrophage-mediated checkpoint system on the survival of patients with cHL.
Collapse
|
25
|
Pollari M, Leivonen SK, Leppä S. Testicular Diffuse Large B-Cell Lymphoma-Clinical, Molecular, and Immunological Features. Cancers (Basel) 2021; 13:cancers13164049. [PMID: 34439203 PMCID: PMC8392512 DOI: 10.3390/cancers13164049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Testicular diffuse large B-cell lymphoma (T-DLBCL) is a rare and aggressive lymphoma entity that mainly affects elderly men. It has a high relapse rate with especially the relapses of the central nervous system associating with dismal outcome. T-DLBCL has a unique biology with distinct genetic characteristics and clinical presentation, and the increasing knowledge on the tumor microenvironment of T-DLBCL highlights the significance of the host immunity and immune escape in this rare lymphoma, presenting in an immune-privileged site of the testis. This review provides an update on the latest progress made in T-DLBCL research and summarizes the clinical perspectives in T-DLBCL. Abstract Primary testicular lymphoma is a rare lymphoma entity, yet it is the most common testicular malignancy among elderly men. The majority of the cases represent non-germinal center B-cell-like (non-GCB) diffuse large B-cell lymphoma (DLBCL) with aggressive clinical behavior and a relatively high relapse rate. Due to the rareness of the disease, no randomized clinical trials have been conducted and the currently recognized standard of care is based on retrospective analyses and few phase II trials. During recent years, the tumor microenvironment (TME) and tumor-related immunity have been the focus of many tumor biology studies, and the emergence of targeted therapies and checkpoint inhibitors has significantly modulated the field of cancer therapies. Testicular DLBCL (T-DLBCL) is presented in an immune-privileged site of the testis, and the roles of NF-κB pathway signaling, 9p24.1 aberrations, and tumor-infiltrating immune cells, especially immune checkpoint expressing lymphocytes and macrophages, seem to be unique compared to other lymphoma entities. Preliminary data on the use of immune checkpoint inhibitors in the treatment of T-DLBCL are promising and more studies are ongoing.
Collapse
Affiliation(s)
- Marjukka Pollari
- Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.-K.L.); (S.L.)
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, 33521 Tampere, Finland
- Correspondence:
| | - Suvi-Katri Leivonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.-K.L.); (S.L.)
- Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, 00029 Helsinki, Finland
| | - Sirpa Leppä
- Research Program Unit, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (S.-K.L.); (S.L.)
- Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, 00029 Helsinki, Finland
| |
Collapse
|
26
|
Wang Z, Chimenti MS, Strouse C, Weiner GJ. T cells, particularly activated CD4 + cells, maintain anti-CD20-mediated NK cell viability and antibody dependent cellular cytotoxicity. Cancer Immunol Immunother 2021; 71:237-249. [PMID: 34110453 PMCID: PMC8783893 DOI: 10.1007/s00262-021-02976-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022]
Abstract
Anti-CD20 monoclonal antibody (mAb) therapy is a mainstay of therapy for B cell malignancies, however many patients fail to respond or eventually develop resistance. The current understanding of mechanisms responsible for this resistance is limited. When peripheral blood mononuclear cells of healthy donors were cultured with Raji cells for 7 days, rituximab (RTX) induced NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC), enhanced NK cell viability and increased or maintained NK expression of CD56, CD16, CD57 and KIR. T cells, mainly CD4+, mediated these changes in a contact-dependent manner, with local T cell production of IL2 playing a central role. Similar findings were found when autologous B cells were used as target cells demonstrating the need for T cell help was not due to allogenic reaction. Results with other anti-CD20 and anti-EGFR antibodies were consistent. Small numbers of T cells activated by anti-CD3/CD28 beads or bispecific antibody enhanced RTX-mediated NK cell ADCC, viability and phenotypical changes. Pathway analysis of bulk NK cell mRNA sequencing after activation by RTX with and without T cells was consistent with T cells maintaining the viability of the activated NK cells. These findings suggest T cell help, mediated in large part by local production of IL2, contributes to NK cell ADCC and viability, and that activating T cells in the tumor microenvironment, such as through the use of anti-CD3 based bispecific antibodies, could enhance the efficacy of anti-CD20 and other mAb therapies where NK-mediated ADCC is a primary mechanism of action.
Collapse
Affiliation(s)
- Zhaoming Wang
- Cancer Biology Graduate Program, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Michael S Chimenti
- Iowa Institute of Human Genetics, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Christopher Strouse
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA
| | - George J Weiner
- Cancer Biology Graduate Program, Holden Comprehensive Cancer Center, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
27
|
Laddaga FE, Ingravallo G, Mestice A, Tamma R, Perrone T, Maiorano E, Ribatti D, Specchia G, Gaudio F. Correlation between circulating blood and microenvironment T lymphocytes in diffuse large B-cell lymphomas. J Clin Pathol 2021; 75:493-497. [PMID: 34011621 DOI: 10.1136/jclinpath-2020-207048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022]
Abstract
AIMS Diffuse large B-cell lymphoma (DLBCL) is characterised by marked clinical and biological heterogeneity, attributable to the tumour cells and their microenvironment. METHODS In this study, we investigated circulating subsets of blood lymphocytes and monocytes and their relationship with T cells in the tumour microenvironment (TME) in chemoresistant and chemosensitive patients with DLBCL. RESULTS The study showed that (1) absolute lymphocyte count (ALC) and CD3+ and CD4+ cells were reduced in chemoresistant patients compared with chemosensitive patients; (2) lymphocyte:monocyte ratio (LMR) showed a positive correlation with peripheral blood CD3+ and CD4+ cells; (3) ALC, LMR, peripheral blood CD3+ and CD4+ cells showed a positive correlation with T cells in the TME. CONCLUSIONS Overall, these data suggest that DLBCL with high TME T cells display a pre-existing antitumour immune response. In the rituximab-containing regimen, TME T cells are stimulated further to participate in the immune response against lymphoma cells. In contrast, DLBCL lymphomas with low T-cell infiltration reflect the absence of a pre-existing antitumour immunity and have a lower likelihood of obtaining an optimal response to therapy.
Collapse
Affiliation(s)
| | | | - Anna Mestice
- Hematology and Stem Cell Transplantation Unit, AOU Consorziale Policlinico, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari, Bari, Italy
| | - Tommasina Perrone
- Hematology and Stem Cell Transplantation Unit, AOU Consorziale Policlinico, Bari, Italy
| | | | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari, Bari, Italy
| | | | - Francesco Gaudio
- Hematology and Stem Cell Transplantation Unit, AOU Consorziale Policlinico, Bari, Italy
| |
Collapse
|
28
|
Autio M, Leivonen SK, Brück O, Mustjoki S, Mészáros Jørgensen J, Karjalainen-Lindsberg ML, Beiske K, Holte H, Pellinen T, Leppä S. Immune cell constitution in the tumor microenvironment predicts the outcome in diffuse large B-cell lymphoma. Haematologica 2021; 106:718-729. [PMID: 32079690 PMCID: PMC7927991 DOI: 10.3324/haematol.2019.243626] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) and limited immune surveillance
play important roles in lymphoma pathogenesis. Here we
aimed to characterize immunological profiles of diffuse large B-cell
lymphoma (DLBCL) and predict the outcome in response to
immunochemotherapy. We profiled the expression of 730 immune-related
genes in tumor tissues of 81 patients with DLBCL utilizing the Nanostring
platform, and used multiplex immunohistochemistry to characterize T-cell
phenotypes, including cytotoxic T cells (CD8, Granzyme B, OX40, Ki67),
T-cell immune checkpoint (CD3, CD4, CD8, PD1, TIM3, LAG3), as well as
regulatory T-cells and Th1 effector cells (CD3, CD4, FOXP3, TBET) in 188
patients. We observed a high degree of heterogeneity at the transcriptome
level. Correlation matrix analysis identified gene expression signatures
with highly correlating genes, the main cluster containing genes for cytolytic
factors, immune checkpoint molecules, T cells and macrophages, together
named a TME immune cell signature. Immunophenotyping of the distinct
cell subsets revealed that a high proportion of immune checkpoint positive
T cells translated to unfavorable survival. Together, our results demonstrate
that the immunological profile of DLBCL TME is heterogeneous and clinically
meaningful. This highlights the potential impact of T-cell immune
checkpoint in regulating survival and resistance to immunochemotherapy.
(Registered at clinicaltrials.gov identifiers: NCT01502982 and NCT01325194.)
Collapse
Affiliation(s)
- Matias Autio
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Suvi-Katri Leivonen
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Oscar Brück
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland,Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Satu Mustjoki
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland,Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland,Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | | | | | - Klaus Beiske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Harald Holte
- Department of Oncology, and KG Jebsen Centre for B Cell Malignancies, Oslo University Hospital, Oslo, Norway
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | - Sirpa Leppä
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland,Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
29
|
Alame M, Cornillot E, Cacheux V, Rigau V, Costes-Martineau V, Lacheretz-Szablewski V, Colinge J. The immune contexture of primary central nervous system diffuse large B cell lymphoma associates with patient survival and specific cell signaling. Am J Cancer Res 2021; 11:3565-3579. [PMID: 33664848 PMCID: PMC7914352 DOI: 10.7150/thno.54343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022] Open
Abstract
Rationale: Primary central nervous system diffuse large B-cell lymphoma (PCNSL) is a rare and aggressive entity that resides in an immune-privileged site. The tumor microenvironment (TME) and the disruption of the immune surveillance influence lymphoma pathogenesis and immunotherapy resistance. Despite growing knowledge on heterogeneous therapeutic responses, no comprehensive description of the PCNSL TME is available. We hence investigated the immune subtypes of PCNSL and their association with molecular signaling and survival. Methods: Analysis of PCNSL transcriptomes (sequencing, n = 20; microarrays, n = 34). Integrated correlation analysis and signaling pathway topology enabled us to infer intercellular interactions. Immunohistopathology and digital imaging were used to validate bioinformatic results. Results: Transcriptomics revealed three immune subtypes: immune-rich, poor, and intermediate. The immune-rich subtype was associated to better survival and characterized by hyper-activation of STAT3 signaling and inflammatory signaling, e.g., IFNγ and TNF-α, resembling the hot subtype described in primary testicular lymphoma and solid cancer. WNT/β-catenin, HIPPO, and NOTCH signaling were hyper-activated in the immune-poor subtype. HLA down-modulation was clearly associated with a low or intermediate immune infiltration and the absence of T-cell activation. Moreover, HLA class I down-regulation was also correlated with worse survival with implications on immune-intermediate PCNSL that frequently feature reduced HLA expression. A ligand-receptor intercellular network revealed high expression of two immune checkpoints, i.e., CTLA-4/CD86 and TIM-3/LAGLS9. TIM-3 and galectin-9 proteins were clearly upregulated in PCNSL. Conclusion: Altogether, our study reveals that patient stratification according to immune subtypes, HLA status, and immune checkpoint molecule quantification should be considered prior to immune checkpoint inhibitor therapy. Moreover, TIM-3 protein should be considered an axis for future therapeutic development.
Collapse
|
30
|
Soluble PD-1 but Not PD-L1 Levels Predict Poor Outcome in Patients with High-Risk Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2021; 13:cancers13030398. [PMID: 33499013 PMCID: PMC7865236 DOI: 10.3390/cancers13030398] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Soluble forms of checkpoint protein PD-1 and its ligand PD-L1 can be measured from circulation, but their source, function, and clinical impact in cancer remain incompletely understood. In this study, we used serum samples collected during a conduction of a prospective immunochemotherapy trial in patients with high-risk diffuse large B-cell lymphoma (DLBCL) and assessed their clinical significance. Our results demonstrate that sPD-1 levels in the peripheral blood at the time of diagnosis correlate with the quantities of tumor infiltrating PD1+ T cells and translate to inferior survival. To our knowledge, this is the first study to identify sPD-1 as a prognostic factor, providing interesting perspectives on future clinical trials in DLBCL, including patients’ stratification associated with checkpoint blockade. Abstract Interaction of checkpoint receptor programmed death 1 (PD-1) with its ligand 1 (PD-L1) downregulates T cell effector functions and thereby leads to tumor immune escape. Here, we aimed to determine the clinical significance of soluble PD-1 (sPD-1) and soluble PD-L1 (sPD-L1) in patients with diffuse large B-cell lymphoma (DLBCL). We included 121 high-risk DLBCL patients treated in the Nordic NLG-LBC-05 trial with dose-dense immunochemotherapy. sPD-1 and sPD-L1 levels were measured from serum samples collected prior to treatment, after three immunochemotherapy courses, and at the end of therapy. sPD-1 and sPD-L1 levels were the highest in pretreatment samples, declining after three courses, and remaining low post-treatment. Pretreatment sPD-1 levels correlated with the quantities of PD1+ T cells in tumor tissue and translated to inferior survival, while no correlation was observed between sPD-L1 levels and outcome. The relative risk of death was 2.9-fold (95% CI 1.12–7.75, p = 0.028) and the risk of progression was 2.8-fold (95% CI 1.16–6.56, p = 0.021) in patients with high pretreatment sPD-1 levels compared to those with low levels. In conclusion, pretreatment sPD-1 level is a predictor of poor outcome after dose-dense immunochemotherapy and may be helpful in further improving molecular risk profiles in DLBCL.
Collapse
|
31
|
Brück O, Lee MH, Turkki R, Uski I, Penttilä P, Paavolainen L, Kovanen P, Järvinen P, Bono P, Pellinen T, Mustjoki S, Kreutzman A. Spatial immunoprofiling of the intratumoral and peritumoral tissue of renal cell carcinoma patients. Mod Pathol 2021; 34:2229-2241. [PMID: 34215851 PMCID: PMC8592837 DOI: 10.1038/s41379-021-00864-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 01/02/2023]
Abstract
While the abundance and phenotype of tumor-infiltrating lymphocytes are linked with clinical survival, their spatial coordination and its clinical significance remain unclear. Here, we investigated the immune profile of intratumoral and peritumoral tissue of clear cell renal cell carcinoma patients (n = 64). We trained a cell classifier to detect lymphocytes from hematoxylin and eosin stained tissue slides. Using unsupervised classification, patients were further classified into immune cold, hot and excluded topographies reflecting lymphocyte abundance and localization. The immune topography distribution was further validated with The Cancer Genome Atlas digital image dataset. We showed association between PBRM1 mutation and immune cold topography, STAG1 mutation and immune hot topography and BAP1 mutation and immune excluded topography. With quantitative multiplex immunohistochemistry we analyzed the expression of 23 lymphocyte markers in intratumoral and peritumoral tissue regions. To study spatial interactions, we developed an algorithm quantifying the proportion of adjacent immune cell pairs and their immunophenotypes. Immune excluded tumors were associated with superior overall survival (HR 0.19, p = 0.02) and less extensive metastasis. Intratumoral T cells were characterized with pronounced expression of immunological activation and exhaustion markers such as granzyme B, PD1, and LAG3. Immune cell interaction occurred most frequently in the intratumoral region and correlated with CD45RO expression. Moreover, high proportion of peritumoral CD45RO+ T cells predicted poor overall survival. In summary, intratumoral and peritumoral tissue regions represent distinct immunospatial profiles and are associated with clinicopathologic characteristics.
Collapse
Affiliation(s)
- Oscar Brück
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland. .,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland. .,Hematology Research Unit Helsinki, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland. .,Comprehensive Cancer Center, Department of Hematology, Helsinki University Hospital, Helsinki, Finland.
| | - Moon Hee Lee
- grid.7737.40000 0004 0410 2071Translational Immunology Research Program, University of Helsinki, Helsinki, Finland ,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland ,grid.15485.3d0000 0000 9950 5666Hematology Research Unit Helsinki, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Riku Turkki
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ilona Uski
- grid.7737.40000 0004 0410 2071Translational Immunology Research Program, University of Helsinki, Helsinki, Finland ,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland ,grid.15485.3d0000 0000 9950 5666Hematology Research Unit Helsinki, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Patrick Penttilä
- grid.15485.3d0000 0000 9950 5666Abdominal Center, Urology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Lassi Paavolainen
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Panu Kovanen
- grid.7737.40000 0004 0410 2071Department of Pathology, HUSLAB, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Petrus Järvinen
- grid.15485.3d0000 0000 9950 5666Abdominal Center, Urology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Petri Bono
- grid.15485.3d0000 0000 9950 5666Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Teijo Pellinen
- grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Satu Mustjoki
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland. .,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland. .,Hematology Research Unit Helsinki, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland. .,Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland.
| | - Anna Kreutzman
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland. .,iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland. .,Hematology Research Unit Helsinki, University of Helsinki and Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
32
|
Menter T, Tzankov A, Dirnhofer S. The tumor microenvironment of lymphomas: Insights into the potential role and modes of actions of checkpoint inhibitors. Hematol Oncol 2020; 39:3-10. [PMID: 33105031 DOI: 10.1002/hon.2821] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment (TME) - a term comprising non-neoplastic cells and extracellular matrix as well as various cytokines, chemokines, growth factors, and other substances in the vicinity of tumor cells - is an integrative part of most tumors including lymphomas. Interactions between lymphoma cells and the TME are vital for survival and proliferation of the former. In addition, lymphoma cells often reprogram the TME to protect them from defense mechanisms of the host's immune system. In this review, we will introduce the role of the tumor microenvironment (TME) for lymphoma cells looking at direct cell-cell interactions as well as cytokine-related communications. The immunomodulative/immunosuppressive role of the TME is more and more coming into the focus of potential new targeted therapies, and thus a special attention will be given to the interactions of immune checkpoints such as programed cell death protein 1 and L1 (PD-1/PD-L1), T-cell immunoglobulin and mucin-domain containing protein-3 (TIM-3), lymphocyte-activation gene 3 (LAG-3), and cytotoxic T-lymphocyte-associated protein-4 (CTLA4) with the TME, as well as their expression by both lymphoma cells and cells of the TME. Aspects of the TME will be discussed for indolent and aggressive B-cell lymphomas, Hodgkin lymphomas, and T-cell lymphomas. In addition, the potential influence of other immunomodulators such as lenalidomide will be briefly touched. The complex role of the TME is in the focus of new therapeutic options. In order to exploit its full therapeutic potential, however, a thorough understanding of TME biology and interaction between lymphoma cells and the TME, as well as the host's immune system and the TME is necessary.
Collapse
Affiliation(s)
- Thomas Menter
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stefan Dirnhofer
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
33
|
Younes S, Natkunam Y. FOXes at play in the lymphoma landscape. Leuk Lymphoma 2020; 62:5-7. [PMID: 33064049 DOI: 10.1080/10428194.2020.1834099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Sheren Younes
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
34
|
Pollari M, Pellinen T, Karjalainen-Lindsberg ML, Kellokumpu-Lehtinen PL, Leivonen SK, Leppä S. Adverse prognostic impact of regulatory T-cells in testicular diffuse large B-cell lymphoma. Eur J Haematol 2020; 105:712-721. [PMID: 32632935 DOI: 10.1111/ejh.13484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Testicular diffuse large B-cell lymphoma (T-DLBCL) is a rare and aggressive extranodal lymphoma. We have previously shown that high content of tumor-infiltrating lymphocytes (TILs) and PD-1 expressing TILs associate with better survival in T-DLBCL. In this study, we have further characterized distinct TIL subtypes and their proportions in association with patient demographics and survival. METHODS We used multiplex immunohistochemistry to characterize TIL phenotypes, including cytotoxic T-cells (CTLs; CD8+ , OX40+ , Granzyme B+ , Ki-67+ , LAG-3+ , TIM-3+ , PD-1+ ), CD4+ T-cells (CD3+ , CD4+ , TIM-3+ , LAG-3+ ), regulatory T-cells (Tregs; CD3+ , CD4+ , FoxP3+ ), and T helper 1 cells (Th1; CD3+ , CD4+ , T-bet+ ) in 79 T-DLBCLs, and correlated the findings with patient demographics and outcome. RESULTS We observed a substantial variation in TIL phenotypes between the patients. The most prominent CD8+ TILs were Ki-67+ and TIM-3+ CTLs, whereas the most prominent CD4+ TILs were FoxP3+ Tregs. Despite the overall favorable prognostic impact of high TIL content, we found a subpopulation of T-bet+ FoxP3+ Tregs that had a significant adverse impact on survival. Lower content of CTLs with activated or exhausted phenotypes correlated with aggressive clinical features. CONCLUSIONS Our results demonstrate significant variation in TIL phenotypes and emphasize the adverse prognostic impact of Tregs in T-DLBCL.
Collapse
Affiliation(s)
- Marjukka Pollari
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), Helsinki, Finland
| | | | - Pirkko-Liisa Kellokumpu-Lehtinen
- Department of Oncology, Tays Cancer Center, Tampere University Hospital, Tampere, Finland.,Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Suvi-Katri Leivonen
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Sirpa Leppä
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland.,iCAN, Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| |
Collapse
|
35
|
Low Absolute Lymphocyte Counts in the Peripheral Blood Predict Inferior Survival and Improve the International Prognostic Index in Testicular Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2020; 12:cancers12071967. [PMID: 32698344 PMCID: PMC7409117 DOI: 10.3390/cancers12071967] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
Low absolute lymphocyte counts (ALC) and high absolute monocyte counts (AMC) are associated with poor survival in patients with diffuse large B-cell lymphoma (DLBCL). We studied the prognostic impact of the ALC and AMC in patients with testicular DLBCL (T-DLBCL). T-DLBCL patients were searched using Southern Finland University Hospital databases and the Danish lymphoma registry. The progression free survival (PFS) and overall survival (OS) were assessed using Kaplan-Meier and Cox proportional hazards methods. We identified 178 T-DLBCL patients, of whom 78 (44%) had a low ALC at diagnosis. The ALC did not correlate with survival in the whole cohort. However, among the patients treated with rituximab (R) containing regimen, a pre-therapeutic low ALC was associated with an increased risk of progression (HR 1.976, 95% CI 1.267–3.086, p = 0.003). Conversely, intravenous (iv) CNS directed chemotherapy translated to favorable outcome. In multivariate analyses, the advantage of an iv CNS directed chemotherapy was sustained (PFS, HR 0.364, 95% CI 0.175–0.757, p = 0.007). The benefit of R and intravenous CNS directed chemotherapy was observed only in non-lymphopenic patients. The AMC did not correlate with survival. A low ALC is an adverse prognostic factor in patients with T-DLBCL. Alternative treatment options for lymphopenic patients are needed.
Collapse
|
36
|
Wudhikarn K, Bunworasate U, Julamanee J, Lekhakula A, Ekwattanakit S, Khuhapinant A, Niparuck P, Chuncharunee S, Numbenjapon T, Prayongratana K, Kanitsap N, Wongkhantee S, Makruasi N, Wong P, Norasetthada L, Nawarawong W, Sirijerachai C, Chansung K, Suwanban T, Praditsuktavorn P, Intragumtornchai T. Event-free survival at 12 months is a strong surrogate endpoint for stage 1 diffuse large B cell lymphoma: a report from Nation Wide Registry Thai Lymphoma Study Group. Leuk Lymphoma 2020; 61:2614-2621. [PMID: 32573294 DOI: 10.1080/10428194.2020.1780586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Event-free survival at 12 months (EFS12) is a surrogate endpoint for long-term outcomes in many histologic lymphoma subtypes. However, most reports have primarily investigated the implication of EFS12 in advanced-stage non-Hodgkin lymphoma (NHL). There are limited data regarding the significance of EFS12 in early-stage NHL. Herein, we evaluated the prognostic significance of EFS12 in patients with stage 1 diffuse large B-cell lymphoma (DLBCL). Out of 282 patients with stage 1 DLBCL who received intensive therapy, 227 (80.5%) achieved EFS12. The 4-year overall survival (OS) was 91.4% and 4.0% for patients who achieved and failed to achieve EFS12, respectively. Multivariable analyses demonstrated response to treatment and achievement of EFS12 as independent predictors for OS. In conclusion, our study demonstrated EFS12 as a powerful prognostic factor for stage 1 DLBCL. Further validation in more extensive prospective studies is warranted.
Collapse
Affiliation(s)
- Kitsada Wudhikarn
- Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Udomsak Bunworasate
- Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Research Unit in Translational Hematology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jakrawadee Julamanee
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Arnuparp Lekhakula
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Supachai Ekwattanakit
- Department of Internal Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Archrob Khuhapinant
- Department of Internal Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pimjai Niparuck
- Department of Internal Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Suporn Chuncharunee
- Department of Internal Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tontanai Numbenjapon
- Department of Internal Medicine, Faculty of Medicine, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Kannadit Prayongratana
- Department of Internal Medicine, Faculty of Medicine, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Nonglak Kanitsap
- Department of Internal Medicine, Faculty of Medicine, Thammasat University, Bangkok, Thailand
| | - Somchai Wongkhantee
- Department of Internal Medicine, Khonkaen Regional Hospital, Khon Kaen, Thailand
| | - Nisa Makruasi
- Department of Internal Medicine, Faculty of Medicine, Srinakharinwirot University, Ongkharak, Thailand
| | - Peerapon Wong
- Department of Internal Medicine, Faculty of Medicine, Naresuan University, Phitsanulok, Thailand
| | - Lalita Norasetthada
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Weerasak Nawarawong
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chittima Sirijerachai
- Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanchana Chansung
- Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Pannee Praditsuktavorn
- Department of Internal Medicine, Chulabhorn Hospital, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Tanin Intragumtornchai
- Department of Internal Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
37
|
Bastid J, Dejou C, Docquier A, Bonnefoy N. The Emerging Role of the IL-17B/IL-17RB Pathway in Cancer. Front Immunol 2020; 11:718. [PMID: 32373132 PMCID: PMC7186465 DOI: 10.3389/fimmu.2020.00718] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 03/30/2020] [Indexed: 12/20/2022] Open
Abstract
Among inflammatory mediators, a growing body of evidence emphasizes the contribution of the interleukin 17 (IL-17) cytokine family in malignant diseases. Besides IL-17A, the prototypic member of the IL-17 family, several experimental findings strongly support the role of the IL-17B/IL-17 receptor B (IL-17RB) pathway in tumorigenesis and resistance to anticancer therapies. In mouse models, IL-17B signaling through IL-17RB directly promotes cancer cell survival, proliferation, and migration, and induces resistance to conventional chemotherapeutic agents. Importantly, recent work by our and other laboratories showed that IL-17B signaling dramatically alters the tumor microenvironment by promoting chemokine and cytokine secretion which foster tumor progression. Moreover, the finding that elevated IL-17B is associated with poor prognosis in patients with pancreatic, gastric, lung, and breast cancer strengthens the results obtained in pre-clinical studies and highlights its clinical relevance. Here, we review the current understanding on the IL-17B/IL-17RB expression patterns and biological activities in cancer and highlight issues that remain to be addressed to better characterize IL-17B and its receptor as potential targets for enhancing the effectiveness of the existing cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Nathalie Bonnefoy
- IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier, Montpellier, France
| |
Collapse
|
38
|
Karihtala K, Leivonen SK, Brück O, Karjalainen-Lindsberg ML, Mustjoki S, Pellinen T, Leppä S. Prognostic Impact of Tumor-Associated Macrophages on Survival Is Checkpoint Dependent in Classical Hodgkin Lymphoma. Cancers (Basel) 2020; 12:cancers12040877. [PMID: 32260340 PMCID: PMC7225916 DOI: 10.3390/cancers12040877] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/25/2020] [Accepted: 03/30/2020] [Indexed: 12/25/2022] Open
Abstract
Tumor microenvironment and immune escape affect pathogenesis and survival in classical Hodgkin lymphoma (cHL). While tumor-associated macrophage (TAM) content has been associated with poor outcomes, macrophage-derived determinants with clinical impact have remained undefined. Here, we have used multiplex immunohistochemistry and digital image analysis to characterize TAM immunophenotypes with regard to expression of checkpoint molecules programmed cell death ligand 1 (PD-L1) and indoleamine 2,3-dioxygenase 1 (IDO-1) from the diagnostic tumor tissue samples of 130 cHL patients, and correlated the findings with clinical characteristics and survival. We show that a large proportion of TAMs express PD-L1 (CD68+, median 32%; M2 type CD163+, median 22%), whereas the proportion of TAMs expressing IDO-1 is lower (CD68+, median 5.5%; CD163+, median 1.4%). A high proportion of PD-L1 and IDO-1 expressing TAMs from all TAMs (CD68+), or from CD163+ TAMs, is associated with inferior outcome. In multivariate analysis with age and stage, high proportions of PD-L1+ and IDO-1+ TAMs remain independent prognostic factors for freedom from treatment failure (PD-L1+CD68+/CD68+, HR = 2.63, 95% CI 1.17–5.88, p = 0.019; IDO-1+CD68+/CD68+, HR = 2.48, 95% CI 1.03–5.95, p = 0.042). In contrast, proportions of PD-L1+ tumor cells, all TAMs or PD-L1− and IDO-1− TAMs are not associated with outcome. The findings implicate that adverse prognostic impact of TAMs is checkpoint-dependent in cHL.
Collapse
Affiliation(s)
- Kristiina Karihtala
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.K.); (S.-K.L.)
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, 00029 Helsinki, Finland
| | - Suvi-Katri Leivonen
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.K.); (S.-K.L.)
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, 00029 Helsinki, Finland
| | - Oscar Brück
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; (O.B.); (S.M.)
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00029 Helsinki, Finland
| | | | - Satu Mustjoki
- Translational Immunology Research Program and Department of Clinical Chemistry and Hematology, University of Helsinki, 00014 Helsinki, Finland; (O.B.); (S.M.)
- Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, 00029 Helsinki, Finland
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland (FIMM), 00014 Helsinki, Finland;
| | - Sirpa Leppä
- Applied Tumor Genomics Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.K.); (S.-K.L.)
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, 00029 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-427-0820
| |
Collapse
|
39
|
Alame M, Pirel M, Costes-Martineau V, Bauchet L, Fabbro M, Tourneret A, De Oliveira L, Durand L, Roger P, Gonzalez S, Cacheux V, Rigau V, Szablewski V. Characterisation of tumour microenvironment and immune checkpoints in primary central nervous system diffuse large B cell lymphomas. Virchows Arch 2019; 476:891-902. [PMID: 31811434 DOI: 10.1007/s00428-019-02695-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022]
Abstract
Primary central nervous system diffuse large B cell lymphoma (PCNS-DLBCL) is a rare and aggressive entity of diffuse large B cell lymphoma (DLBCL). Elements of the tumour microenvironment (TME) including tumour-infiltrating lymphocytes (TILs) and tumour-associated macrophages (TAMs) have been associated with survival in DLBCL but their composition and prognostic impact in PCNS-DLBCL are unknown. Programmed cell death-1 (PD1)/programmed death-ligand 1 (PD-L1) immune checkpoint may represent a therapeutic option. Here, we aimed to characterise PD1/PDL1 immune checkpoints and the composition of the TME in PCNS-DLBCL. We collected tumour tissue and clinical data from 57 PCNS-DLBCL and used immunohistochemistry to examine TAMs (CD68, CD163), TILs (CD3, CD4, CD8, PD1) and tumour B cells (PAX5/PDL1 double stains, PDL1). The PDL1 gene was evaluated by fluorescence in situ hybridization (FISH). PAX5/PDL1 identified PDL1 expression by tumour B cells in 10/57 cases (17.5%). PDL1 gene translocation was a recurrent cytogenetic alteration in PNCS-DLBCL (8/47.17%) and was correlated with PDL1 positive expression in tumour B cells. The TME consisted predominantly of CD163 (+) M2 TAMs and CD8 (+) TILs. Most TAMs expressed PDL1 and most TILs expressed PD1. The density of TAMs and TILs did not associate with outcome. We showed that expression of PD1 on TILs and PDL1 on TAMs, but not the expression of PDL1 on tumour B cells was correlated with better prognosis. These findings support a significant role of TME composition and PD1/PDL1 crosstalk in PCNS-DLBCL pathogenesis and bring new insights to the targeted therapy of this aggressive lymphoma.
Collapse
Affiliation(s)
- Melissa Alame
- Département d'Hématologie biologique, CHU Montpellier, Hôpital Saint Eloi, 34275, Montpellier, France.,Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France
| | - Marion Pirel
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Biopathologie, CHU Montpellier, Hôpital Gui De Chauliac, 34295, Montpellier, France
| | - Valérie Costes-Martineau
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Biopathologie, CHU Montpellier, Hôpital Gui De Chauliac, 34295, Montpellier, France
| | - Luc Bauchet
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Neurochirurgie, CHU Montpellier, Hôpital Gui De Chauliac, 34000, Montpellier, France
| | - Michel Fabbro
- Département d'Oncologie Médicale, Institut du Cancer de Montpellier, Parc Euromédecine, 208 rue des Apothicaires, 34298, Montpellier, France
| | - Alicia Tourneret
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Biopathologie, CHU Montpellier, Hôpital Gui De Chauliac, 34295, Montpellier, France
| | - Laura De Oliveira
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Biopathologie, CHU Montpellier, Hôpital Gui De Chauliac, 34295, Montpellier, France
| | | | - Pascal Roger
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Biopathologie, CHU Nîmes, Hôpital Caremeau, 30029, Nîmes, France
| | - Samia Gonzalez
- Département de Biopathologie, CHU Nîmes, Hôpital Caremeau, 30029, Nîmes, France
| | - Valère Cacheux
- Département d'Hématologie biologique, CHU Montpellier, Hôpital Saint Eloi, 34275, Montpellier, France.,Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France
| | - Valérie Rigau
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France.,Département de Biopathologie, CHU Montpellier, Hôpital Gui De Chauliac, 34295, Montpellier, France
| | - Vanessa Szablewski
- Faculté de Médecine Montpellier Nîmes, 2 rue école de Médecine, 34060, Montpellier, France. .,Département de Biopathologie, CHU Montpellier, Hôpital Gui De Chauliac, 34295, Montpellier, France.
| |
Collapse
|
40
|
Mulder TA, Wahlin BE, Österborg A, Palma M. Targeting the Immune Microenvironment in Lymphomas of B-Cell Origin: From Biology to Clinical Application. Cancers (Basel) 2019; 11:cancers11070915. [PMID: 31261914 PMCID: PMC6678966 DOI: 10.3390/cancers11070915] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/08/2023] Open
Abstract
In lymphomas of B-cell origin, cancer cells orchestrate an inflammatory microenvironment of immune and stromal cells that sustain the tumor cell survival and growth, known as a tumor microenvironment (TME). The features of the TME differ between the different lymphoma types, ranging from extremely inflammatory, such as in Hodgkin lymphoma, to anergic, leading to immune deficiency and susceptibility to infections, such as in chronic lymphocytic leukemia. Understanding the characteristic features of the TME as well as the interactions between cancer and TME cells has given insight into the pathogenesis of most lymphomas and contributed to identify novel therapeutic targets. Here, we summarize the preclinical data that contributed to clarifying the role of the immune cells in the TME of different types of lymphomas of B-cell origin, and explain how the understanding of the biological background has led to new clinical applications. Moreover, we provide an overview of the clinical results of trials that assessed the safety and efficacy of drugs directly targeting TME immune cells in lymphoma patients.
Collapse
Affiliation(s)
- Tom A Mulder
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Björn E Wahlin
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Österborg
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Marzia Palma
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
41
|
Sorenson L, Fu Y, Hood T, Warren S, McEachron TA. Targeted transcriptional profiling of the tumor microenvironment reveals lymphocyte exclusion and vascular dysfunction in metastatic osteosarcoma. Oncoimmunology 2019; 8:e1629779. [PMID: 31428529 PMCID: PMC6685511 DOI: 10.1080/2162402x.2019.1629779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/04/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone tumor in pediatric and adolescent/young adult patients yet little is known about the microenvironment that supports this aggressive disease. We have used targeted gene expression profiling and immunohistochemistry to characterize the microenvironment of metastatic and non-metastatic OS specimens from pediatric patients exhibiting poor histologic response to chemotherapy. Our results indicate that metastatic specimens exhibit lymphocyte exclusion as T cells are confined to the periphery of the pulmonary lesions. Furthermore, our data provides evidence of vascular dysfunction in metastatic OS indicated by increased expression of VEGFA, an increased ANGPT2:ANGPT1 gene expression ratio, and decreased expression of SELE, the gene encoding the adhesion molecule E-selectin. Moreover, correlation analyses show an inverse relationship between lymphocyte abundance and markers of vascular dysfunction exclusively in the metastatic specimens. Together, our data shows that the non-metastatic OS specimens demonstrate increased expression of various immunotherapeutic targets in comparison metastatic specimens and identifies vascular dysfunction and lymphocyte exclusion as important processes for therapeutic intervention in metastatic disease.
Collapse
Affiliation(s)
- Laurie Sorenson
- Department of Translational Genomics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Yanfen Fu
- NanoString Technologies, Inc., Seattle, WA, USA
| | - Tressa Hood
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Troy A. McEachron
- Department of Translational Genomics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
- Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
42
|
Plasma-Derived Reactive Species Shape a Differentiation Profile in Human Monocytes. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9122530] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background: Monocyte-derived macrophages are key regulators and producers of reactive oxygen and nitrogen species (ROS/RNS). Pre-clinical and clinical studies suggest that cold physical plasma may be beneficial in the treatment of inflammatory conditions via the release of ROS/RNS. However, it is unknown how plasma treatment affects monocytes and their differentiation profile. Methods: Naïve or phorbol-12-myristate-13-acetate (PMA)-pulsed THP-1 monocytes were exposed to cold physical plasma. The cells were analyzed regarding their metabolic activity as well as flow cytometry (analysis of viability, oxidation, surface marker expression and cytokine secretion) and high content imaging (quantitative analysis of morphology. Results: The plasma treatment affected THP-1 metabolisms, viability, and morphology. Furthermore, a significant modulation CD55, CD69, CD271 surface-expression and increase of inflammatory IL1β, IL6, IL8, and MCP1 secretion was observed upon plasma treatment. Distinct phenotypical changes in THP-1 cells arguing for a differentiation profile were validated in primary monocytes from donor blood. As a functional outcome, plasma-treated monocytes decreased the viability of co-cultured melanoma cells to a greater extent than their non-treated counterparts. Conclusions: Our results suggest plasma-derived ROS/RNS shaped a differentiation profile in human monocytes as evidenced by their increased inflammatory profile (surface marker and cytokines) as well as functional outcome (tumor toxicity).
Collapse
|
43
|
Mannisto S, Vähämurto P, Pollari M, Clausen MR, Jyrkkiö S, Kellokumpu-Lehtinen PL, Kovanen P, Karjalainen-Lindsberg ML, d'Amore F, Leppä S. Intravenous but not intrathecal central nervous system-directed chemotherapy improves survival in patients with testicular diffuse large B-cell lymphoma. Eur J Cancer 2019; 115:27-36. [PMID: 31082690 DOI: 10.1016/j.ejca.2019.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/23/2019] [Accepted: 04/02/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Testicular lymphoma is a rare malignancy affecting mainly elderly men, the majority representing diffuse large B-cell lymphoma (DLBCL). Its relapse rate is higher than that of nodal DLBCL, often affecting the central nervous system (CNS) with dismal prognosis. PATIENTS AND METHODS We searched for patients with testicular DLBCL (T-DLBCL) involvement from the pathology databases of Southern Finland University Hospitals and the Danish Lymphoma Registry. Clinical information was collected, and outcomes between treatment modalities were evaluated. Progression-free survival (PFS), disease-specific survival (DSS) and overall survival (OS) were assessed using Kaplan-Meier and Cox proportional hazards methods. RESULTS We identified 235 patients; of whom, 192 were treated with curative anthracycline-based chemotherapy. Full survival data were available for 189 patients. In univariate analysis, intravenous CNS-directed chemotherapy, and irradiation or orchiectomy of the contralateral testis translated into favourable PFS, DSS and OS, particularly among the elderly patients (each p ≤ 0.023). Intrathecal chemotherapy had no impact outcome. In multivariate analyses, the advantage of intravenous CNS-directed chemotherapy (hazard ration [HR] for OS, 0.419; 95% confidence interval [CI], 0.256-0.686; p = 0.001) and prophylactic treatment of contralateral testis (HR for OS, 0.514; 95% CI, 0.338-0.782; p = 0.002) was maintained. Rituximab improved survival only among high-risk patients (International Prognostic Index≥3, p = 0.019). The cumulative risk of CNS progression was 8.4% and did not differ between treatment modalities. CONCLUSION The results support the use of CNS-directed chemotherapy and prophylactic treatment of the contralateral testis in patients with T-DLBCL involvement. Survival benefit appears resulting from better control of systemic disease rather than prevention of CNS progression.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Central Nervous System Neoplasms/mortality
- Central Nervous System Neoplasms/prevention & control
- Central Nervous System Neoplasms/secondary
- Databases, Factual
- Denmark
- Disease Progression
- Finland
- Humans
- Infusions, Intravenous
- Infusions, Spinal
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/radiotherapy
- Male
- Middle Aged
- Orchiectomy
- Progression-Free Survival
- Registries
- Risk Assessment
- Risk Factors
- Testicular Neoplasms/drug therapy
- Testicular Neoplasms/mortality
- Testicular Neoplasms/pathology
- Testicular Neoplasms/radiotherapy
- Time Factors
Collapse
Affiliation(s)
- S Mannisto
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - P Vähämurto
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - M Pollari
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Oncology, Tampere University Hospital and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - M R Clausen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - S Jyrkkiö
- Department of Oncology and Radiotherapy, Turku University Central Hospital, Turku, Finland
| | - P-L Kellokumpu-Lehtinen
- Department of Oncology, Tampere University Hospital and Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - P Kovanen
- Department of Pathology, Helsinki University Hospital, Helsinki, Finland
| | | | - F d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - S Leppä
- Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|