1
|
Linton KM, Specht L, Pavlovsky A, Thompson CA, Kimby E, de Jong D, Nastoupil LJ, Cottereau AS, Casulo C, Sarkozy C, Okosun J. Personalised therapy in follicular lymphoma - is the dial turning? Hematol Oncol 2024; 42:e3205. [PMID: 37482955 DOI: 10.1002/hon.3205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/25/2023] [Accepted: 06/17/2023] [Indexed: 07/25/2023]
Abstract
Follicular lymphoma is the most common indolent lymphoma accounting for approximately 20%-25% of all new non-Hodgkin lymphoma diagnoses in western countries. Whilst outcomes are mostly favorable, the spectrum of clinical phenotypes includes high-risk groups with significantly inferior outcomes. This review discusses recent updates in risk stratification and treatment approaches from upfront treatment for limited and advanced stage follicular lymphoma to the growing options for relapsed, refractory disease with perspectives on how to approach this from a personalized lens. Notable gaps remain on how one can precisely and prospectively select optimal treatment for patients based on varying risks, with an anticipation that an increased understanding of the biology of these different phenotypes and increasing refinement of imaging- and biomarker-based tools will, in time, allow these gaps to be closed.
Collapse
Affiliation(s)
- Kim M Linton
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Lena Specht
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Astrid Pavlovsky
- Department of Hematology, Fundaleu Clinical Research Center, Buenos Aires, Argentina
- Centro de Helmatología Pavlovsky, Medical Director, Buenos Aires, Argentina
| | - Carrie A Thompson
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eva Kimby
- Department of Medicine Karolinska Institutet, Center of Hematology, Stockholm, Sweden
| | - Daphne de Jong
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Loretta J Nastoupil
- Department of Lymphoma/Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anne-Ségolène Cottereau
- Department of Nuclear Medicine, Cochin Hospital, APHP, University of Paris Cité, Paris, France
| | - Carla Casulo
- Department of Medicine, University of Rochester, Rochester, New York, USA
| | | | - Jessica Okosun
- Centre for Haemato-Oncology Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
2
|
Collin M, Gagey G, Shanmugam V, Louissaint A, Okosun J, Sarkozy C, Nadel B. Follicular lymphoma research: an open dialogue for a collaborative roadmap. Histopathology 2024. [PMID: 39468961 DOI: 10.1111/his.15344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/30/2024]
Abstract
Follicular lymphoma (FL) is the second most common type of lymphoma (20% of all non-Hodgkin lymphomas), derived from germinal centre (GC) B cells, and is characterised by its significant clinical, prognostic and biological heterogeneity, leading to complexity in management. Despite significant biological investigation and indisputable clinical progress since the advent of the immunotherapy era more than 20 years ago, much remains to be done to understand and cure this lymphoma. Today, FL is metaphorically a giant puzzle on the table with patches of sky, landscape and foliage clearly appearing. However, many of the remaining pieces are held by various stakeholders (e.g. clinicians, pathologists, researchers, drug developers) without global agreement on what the gaps are, or any clear blueprint on how to solve the puzzle of understanding the heterogeneity of this disease and create curative and tailored therapies. With the advent of new investigation and drug technologies, together with recent advances in our capacity to manage big data, the time seems ripe for a change of scale. More than ever, this will require collaboration between and within all stakeholders to overcome the current bottlenecks in the field. As for every investigator, we acknowledge that this first draft is necessarily biased, incomplete and some FL expert readers might recognise some remaining gaps not addressed. We hope they will reply to make this effort a collaborative one to assemble all the pieces in the most ideal fashion. As such, this review intends to be a first step and an interactive platform to a collaborative roadmap towards better understanding and care of FL.
Collapse
Affiliation(s)
- Mélanie Collin
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Guillemette Gagey
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Vignesh Shanmugam
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Abner Louissaint
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Okosun
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Clementine Sarkozy
- Hematology Department, Institut Curie, Saint Cloud, France and LITO, U1288, Université Versailles Saint Quentin en Yveline, Saint Quentin en Yveline, France
| | - Bertrand Nadel
- Aix-Marseille University, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
3
|
Enemark MH, Hemmingsen JK, Jensen ML, Kridel R, Ludvigsen M. Molecular Biomarkers in Prediction of High-Grade Transformation and Outcome in Patients with Follicular Lymphoma: A Comprehensive Systemic Review. Int J Mol Sci 2024; 25:11179. [PMID: 39456961 PMCID: PMC11508793 DOI: 10.3390/ijms252011179] [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: 09/06/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/28/2024] Open
Abstract
Follicular lymphoma (FL) is the most prevalent indolent B-cell lymphoma entity, often characterized by the t(14;18) BCL2-IGH translocation. The malignancy represents a clinically and biologically highly heterogeneous disease. Most patients have favorable prognoses; however, despite therapeutic advancements, the disease remains incurable, with recurrent relapses or early disease progression. Moreover, transformation to an aggressive histology, most often diffuse large-B-cell lymphoma, remains a critical event in the disease course, which is associated with poor outcomes. Understanding the individual patient's risk of transformation remains challenging, which has motivated much research on novel biomarkers within the past four decades. This review systematically assessed the research on molecular biomarkers in FL transformation and outcome. Following the PRISMA guidelines for systemic reviews, the PubMed database was searched for English articles published from January 1984 through September 2024, yielding 6769 results. The identified publications were carefully screened and reviewed, of which 283 original papers met the inclusion criteria. The included studies focused on investigating molecular biomarkers as predictors of transformation or as prognostic markers of time-related endpoints (survival, progression, etc.). The effects of each biomarker were categorized based on their impact on prognosis or risk of transformation as none, favorable, or inferior. The biomarkers included genetic abnormalities, gene expression, microRNAs, markers of B cells/FL tumor cells, markers of the tumor microenvironment, and soluble biomarkers. This comprehensive review provides an overview of the research conducted in the past four decades, underscoring the persistent challenge in risk anticipation of FL patients.
Collapse
Affiliation(s)
- Marie Hairing Enemark
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus N, Denmark; (M.H.E.); (J.K.H.); (M.L.J.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jonas Klejs Hemmingsen
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus N, Denmark; (M.H.E.); (J.K.H.); (M.L.J.)
| | - Maja Lund Jensen
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus N, Denmark; (M.H.E.); (J.K.H.); (M.L.J.)
| | - Robert Kridel
- Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 2C4, Canada;
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, 8200 Aarhus N, Denmark; (M.H.E.); (J.K.H.); (M.L.J.)
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus C, Denmark
| |
Collapse
|
4
|
Narita H, Kuroiwa K, Kawaguchi Y, Murai S, Sasaki Y, Homma M, Kawamata N, Hayashi H, Nagao K, Okamura R, Uesugi Y, Sasaki Y, Shimada S, Watanuki M, Arai N, Yanagisawa K, Shiozawa E, Yamochi T, Hattori N. High Ki-67 Expression Predicting a Risk Factor for the Progression of Disease within 24 Months and Microenvironment in Follicular Lymphoma. Int J Mol Sci 2024; 25:11057. [PMID: 39456838 PMCID: PMC11507466 DOI: 10.3390/ijms252011057] [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: 09/22/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Most follicular lymphomas (FLs) demonstrate an indolent clinical course with favorable outcomes; however, a fraction of patients experiences progression of disease within 24 months (POD24) and has adverse outcomes. This study aimed to determine the predictive risk factors for POD24 in patients with FL, and the characteristics of the microenvironment in FL with POD24. By multivariate analysis, we revealed that increased Ki-67 expression was associated with POD24 events in patients with FL (hazard ratio [HR]: 6.29, 95% confidence interval [CI]: 1.96-20.22, p = 0.0020). Additionally, patients with FL with POD24 demonstrated immune cell reduction by immunohistochemistry analysis. Our results help better understand the therapeutic strategies for FL with POD24.
Collapse
Affiliation(s)
- Hinako Narita
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Kai Kuroiwa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Yukiko Kawaguchi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - So Murai
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (S.M.); (Y.S.); (M.H.); (E.S.); (T.Y.)
| | - Yosuke Sasaki
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (S.M.); (Y.S.); (M.H.); (E.S.); (T.Y.)
| | - Mayumi Homma
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (S.M.); (Y.S.); (M.H.); (E.S.); (T.Y.)
| | - Natsuki Kawamata
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Hidenori Hayashi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Kazuki Nagao
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Reiko Okamura
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Yuka Uesugi
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Yohei Sasaki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Shotaro Shimada
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Megumi Watanuki
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Nana Arai
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Kouji Yanagisawa
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| | - Eisuke Shiozawa
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (S.M.); (Y.S.); (M.H.); (E.S.); (T.Y.)
| | - Toshiko Yamochi
- Department of Pathology and Laboratory Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (S.M.); (Y.S.); (M.H.); (E.S.); (T.Y.)
| | - Norimichi Hattori
- Division of Hematology, Department of Medicine, Showa University School of Medicine, Tokyo 142-8666, Japan; (H.N.); (K.K.); (Y.K.); (N.K.); (H.H.); (K.N.); (R.O.); (Y.U.); (Y.S.); (S.S.); (M.W.); (N.A.); (K.Y.)
| |
Collapse
|
5
|
Zhang S, Wang X, Yang Z, Ding M, Zhang M, Young KH, Zhang X. Minimal residual disease detection in lymphoma: methods, procedures and clinical significance. Front Immunol 2024; 15:1430070. [PMID: 39188727 PMCID: PMC11345172 DOI: 10.3389/fimmu.2024.1430070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
Lymphoma is a highly heterogeneous lymphohematopoietic tumor. As our understanding of the biological and pathological characteristics of lymphoma improves, we are identifying an increasing number of lymphoma subtypes. Genotyping has enhanced our ability to diagnose, treat, and monitor the prognosis of lymphoma. Despite significant improvements in treatment effectiveness, traditional methods for assessing disease response and monitoring prognosis are imperfect, and there is no significant improvement in overall remission rates for lymphoma patients. Minimal Residual Disease (MRD) is often indicative of refractory disease or early relapse. For lymphoma patients, personalized MRD monitoring techniques offer an efficient means to estimate disease remission levels, predict early relapse risk, and assess the effectiveness of new drug regimens. In this review, we delve into the MRD procedures in lymphoma, including sample selection and requirements, detection methods and their limitations and advantages, result interpretation. Besides, we also introduce the clinical applications of MRD detection in lymphoma.
Collapse
Affiliation(s)
- Sijun Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Xiangyu Wang
- School of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Mengjie Ding
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| | - Ken H. Young
- Division of Hematopathology, Duke University Medicine Center, Duke Cancer Institute, Durham, NC, United States
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Lymphoma Diagnosis and Treatment Center of Henan Province, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Wang JN, Zheng G, Wu W, Huang H. Follicular helper T cells: emerging roles in lymphomagenesis. J Leukoc Biol 2024; 116:54-63. [PMID: 37939814 DOI: 10.1093/jleuko/qiad140] [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: 04/29/2023] [Revised: 08/11/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
Follicular helper T cells are a subset of CD4+ T cells that are fundamental to forming germinal centers, which are the primary sites of antibody affinity maturation and the proliferation of activated B cells. Follicular helper T cells have been extensively studied over the past 10 years, especially regarding their roles in cancer genesis. This review describes the characteristics of normal follicular helper T cells and focuses on the emerging link between follicular helper T cells and lymphomagenesis. Advances in lymphoma genetics have substantially expanded our understanding of the role of follicular helper T cells in lymphomagenesis. Moreover, we detail a range of agents and new therapies, with a major focus on chimeric antigen receptor T-cell therapy; these novel approaches may offer new treatment opportunities for patients with lymphomas.
Collapse
Affiliation(s)
- Ji-Nuo Wang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| | - Gaofeng Zheng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| | - Wenjun Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, 311106, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, No.17 Old Zhejiang University Road, Hangzhou, 311112, China
| |
Collapse
|
7
|
Watanabe T, Matsuno Y, Wakabayashi M, Maruyama D, Yamamoto K, Kubota N, Shimada K, Asagoe K, Yamaguchi M, Ando K, Ogura M, Kuroda J, Suehiro Y, Tsukasaki K, Tobinai K, Nagai H. Analyzing the risk factors for disease progression within 2 years and histological transformation in patients treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone as first-line treatment: A 15-year follow-up of patients with advanced follicular lymphoma in JCOG0203. Hematol Oncol 2024; 42:e3272. [PMID: 38595316 DOI: 10.1002/hon.3272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/23/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
Follicular lymphoma (FL) is an indolent lymphoma that becomes aggressive due to histological transformation (HT), leading to reduced survival. Patients with FL have different clinical courses and various treatment options. Some patients exhibit shorter survival and experience disease progression within 24 months of diagnosis/treatment (POD24); the optimal treatment remains an unmet needs. Thus, identifying factors that predict shorter survival is essential to stratify treatment and prolong the survival of patients with FL. To analyze risk factors for POD24 and HT in patients treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) as first-line treatment, we performed this post-hoc analysis of patients with advanced indolent B-cell lymphoma in a randomized clinical trial wherein six cycles of R-CHOP were administered every 2-3 weeks. The primary analysis showed no differences in outcomes, which enabled the analysis of 248 patients with FL, assigned to two arms. All histopathological specimens from the 300 enrolled patients were reviewed by three expert hematopathologists. Multivariable analysis implicated Follicular Lymphoma International Prognostic Index (FLIPI) intermediate (odds ratio [OR] 2.531, 95% confidence interval [CI] 0.676-9.466) and high- (OR 2.236, 95% CI 0.160-31.226) risks, B symptoms (OR 2.091, 95% CI 0.747-5.851), and grade 3A (G3A) (OR 1.833, 95% CI 0.634-5.299) as risk factors for POD24. Furthermore, multivariable analysis through a median follow-up of 15.9 years implicated G3A (OR 2.628, 95% CI 0.806-8.575) and high-risk FLIPI (OR 4.401, 95% CI 0.186-104.377) as risk factors for HT. However, an analysis limited to the first 10 years revealed that the prognostic factors elucidated from the longer-term analysis had a greater impact on HT. G3A and high-risk FLIPI may independently predict POD24 and HT, thereby informing treatment stratification of patients with untreated advanced-stage FL in future trials, particularly to address the unmet needs of patients with POD24.
Collapse
Affiliation(s)
- Takashi Watanabe
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
- Department of Personalized Control Science of Myeloid and Lymphoid Tumors, Mie University Graduate School of Medicine, Tsu, Japan
| | - Yoshihiro Matsuno
- Department of Pathology, National Cancer Center Hospital, Tokyo, Japan
- Pathology Center, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | | | - Dai Maruyama
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
- Department of Hematology Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuhito Yamamoto
- Department of Hematology and Cell Therapy, Aichi Cancer Center, Nagoya, Japan
| | - Nobuko Kubota
- Department of Hematology, Saitama Cancer Center, Saitama, Japan
- Department of Hematology, Shin-Yurigaoka General Hospital, Kawasaki, Japan
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohsuke Asagoe
- Department of Hematology and Oncology, Shiga General Hospital, Moriyama, Japan
| | - Motoko Yamaguchi
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan
- Department of Hematological Malignancies, Mie University Graduate School of Medicine, Tsu, Japan
| | - Kiyoshi Ando
- Division of Hematology/Oncology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Michinori Ogura
- Department of Hematology and Oncology, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Youko Suehiro
- Department of Hematology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Kunihiro Tsukasaki
- Department of Hematology, International Medical Center, Saitama Medical University, Saitama, Japan
| | - Kensei Tobinai
- Department of Hematology, National Cancer Center Hospital, Tokyo, Japan
| | - Hirokazu Nagai
- Department of Hematology and Oncology Research, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
8
|
Laurent C, Dietrich S, Tarte K. Cell cross talk within the lymphoma tumor microenvironment: follicular lymphoma as a paradigm. Blood 2024; 143:1080-1090. [PMID: 38096368 DOI: 10.1182/blood.2023021000] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/30/2023] [Indexed: 03/22/2024] Open
Abstract
ABSTRACT Follicular lymphoma (FL) is an indolent yet incurable germinal center B-cell lymphoma retaining a characteristic follicular architecture. FL tumor B cells are highly dependent on direct and indirect interactions with a specific and complex tumor microenvironment (TME). Recently, great progress has been made in describing the heterogeneity and dynamics of the FL TME and in depicting how tumor clonal and functional heterogeneity rely on the integration of TME-related signals. Specifically, the FL TME is enriched for exhausted cytotoxic T cells, immunosuppressive regulatory T cells of various origins, and follicular helper T cells overexpressing B-cell and TME reprogramming factors. FL stromal cells have also emerged as crucial determinants of tumor growth and remodeling, with a key role in the deregulation of chemokines and extracellular matrix composition. Finally, tumor-associated macrophages play a dual function, contributing to FL cell phagocytosis and FL cell survival through long-lasting B-cell receptor activation. The resulting tumor-permissive niches show additional layers of site-to-site and kinetic heterogeneity, which raise questions about the niche of FL-committed precursor cells supporting early lymphomagenesis, clonal evolution, relapse, and transformation. In turn, FL B-cell genetic and nongenetic determinants drive the reprogramming of FL immune and stromal TME. Therefore, offering a functional picture of the dynamic cross talk between FL cells and TME holds the promise of identifying the mechanisms of therapy resistance, stratifying patients, and developing new therapeutic approaches capable of eradicating FL disease in its different ecosystems.
Collapse
Affiliation(s)
- Camille Laurent
- Department of Pathology, Institut Universitaire du Cancer de Toulouse Oncopole, Centre Hospitalo-Universitaire Toulouse, Centre de Recherches en Cancérologie de Toulouse, Laboratoire d'Excellence TOUCAN, INSERM Unité Mixte de Recherche 1037, Toulouse, France
| | - Sascha Dietrich
- Department of Haematology and Oncology, University Hospital Düsseldorf and Center for Integrated Oncology Aachen Bonn Cologne, Düsseldorf, Germany
| | - Karin Tarte
- Unité Mixte de Recherche S1236, INSERM, Université de Rennes, Etablissement Français du Sang Bretagne, Equipe Labellisée Ligue, Rennes, France
- Department of Biology, Centre Hospitalo-Universitaire de Rennes, Rennes, France
| |
Collapse
|
9
|
Krull JE, Wenzl K, Hopper MA, Manske MK, Sarangi V, Maurer MJ, Larson MC, Mondello P, Yang Z, Novak JP, Serres M, Whitaker KR, Villasboas Bisneto JC, Habermann TM, Witzig TE, Link BK, Rimsza LM, King RL, Ansell SM, Cerhan JR, Novak AJ. Follicular lymphoma B cells exhibit heterogeneous transcriptional states with associated somatic alterations and tumor microenvironments. Cell Rep Med 2024; 5:101443. [PMID: 38428430 PMCID: PMC10983045 DOI: 10.1016/j.xcrm.2024.101443] [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: 09/16/2022] [Revised: 08/14/2023] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Follicular lymphoma (FL) is an indolent non-Hodgkin lymphoma of germinal center origin, which presents with significant biologic and clinical heterogeneity. Using RNA-seq on B cells sorted from 87 FL biopsies, combined with machine-learning approaches, we identify 3 transcriptional states that divide the biological ontology of FL B cells into inflamed, proliferative, and chromatin-modifying states, with relationship to prior GC B cell phenotypes. When integrated with whole-exome sequencing and immune profiling, we find that each state was associated with a combination of mutations in chromatin modifiers, copy-number alterations to TNFAIP3, and T follicular helper cells (Tfh) cell interactions, or primarily by a microenvironment rich in activated T cells. Altogether, these data define FL B cell transcriptional states across a large cohort of patients, contribute to our understanding of FL heterogeneity at the tumor cell level, and provide a foundation for guiding therapeutic intervention.
Collapse
Affiliation(s)
| | - Kerstin Wenzl
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Matthew J Maurer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Melissa C Larson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - ZhiZhang Yang
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | - Brian K Link
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa, Iowa City, IA, USA
| | - Lisa M Rimsza
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Rebecca L King
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
10
|
Stepanenko AA, Sosnovtseva AO, Valikhov MP, Chernysheva AA, Abramova OV, Pavlov KA, Chekhonin VP. Systemic and local immunosuppression in glioblastoma and its prognostic significance. Front Immunol 2024; 15:1326753. [PMID: 38481999 PMCID: PMC10932993 DOI: 10.3389/fimmu.2024.1326753] [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: 10/23/2023] [Accepted: 02/06/2024] [Indexed: 04/07/2024] Open
Abstract
The effectiveness of tumor therapy, especially immunotherapy and oncolytic virotherapy, critically depends on the activity of the host immune cells. However, various local and systemic mechanisms of immunosuppression operate in cancer patients. Tumor-associated immunosuppression involves deregulation of many components of immunity, including a decrease in the number of T lymphocytes (lymphopenia), an increase in the levels or ratios of circulating and tumor-infiltrating immunosuppressive subsets [e.g., macrophages, microglia, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs)], as well as defective functions of subsets of antigen-presenting, helper and effector immune cell due to altered expression of various soluble and membrane proteins (receptors, costimulatory molecules, and cytokines). In this review, we specifically focus on data from patients with glioblastoma/glioma before standard chemoradiotherapy. We discuss glioblastoma-related immunosuppression at baseline and the prognostic significance of different subsets of circulating and tumor-infiltrating immune cells (lymphocytes, CD4+ and CD8+ T cells, Tregs, natural killer (NK) cells, neutrophils, macrophages, MDSCs, and dendritic cells), including neutrophil-to-lymphocyte ratio (NLR), focus on the immune landscape and prognostic significance of isocitrate dehydrogenase (IDH)-mutant gliomas, proneural, classical and mesenchymal molecular subtypes, and highlight the features of immune surveillance in the brain. All attempts to identify a reliable prognostic immune marker in glioblastoma tissue have led to contradictory results, which can be explained, among other things, by the unprecedented level of spatial heterogeneity of the immune infiltrate and the significant phenotypic diversity and (dys)functional states of immune subpopulations. High NLR is one of the most repeatedly confirmed independent prognostic factors for shorter overall survival in patients with glioblastoma and carcinoma, and its combination with other markers of the immune response or systemic inflammation significantly improves the accuracy of prediction; however, more prospective studies are needed to confirm the prognostic/predictive power of NLR. We call for the inclusion of dynamic assessment of NLR and other blood inflammatory markers (e.g., absolute/total lymphocyte count, platelet-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, systemic immune-inflammation index, and systemic immune response index) in all neuro-oncology studies for rigorous evaluation and comparison of their individual and combinatorial prognostic/predictive significance and relative superiority.
Collapse
Affiliation(s)
- Aleksei A. Stepanenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasiia O. Sosnovtseva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marat P. Valikhov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| | - Anastasia A. Chernysheva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Olga V. Abramova
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Konstantin A. Pavlov
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky National Medical Research Center of Psychiatry and Narcology, the Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Medical Nanobiotechnology, Institute of Translational Medicine, N. I. Pirogov Russian National Research Medical University, The Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
11
|
Yang ZZ, Kim HJ, Wu H, Tang X, Yu Y, Krull J, Larson DP, Moore RM, Maurer MJ, Pavelko KD, Jalali S, Pritchett JC, Mudappathi R, Wang J, Villasboas JC, Mondello P, Novak AJ, Ansell SM. T-cell phenotype including CD57 + T follicular helper cells in the tumor microenvironment correlate with a poor outcome in follicular lymphoma. Blood Cancer J 2023; 13:124. [PMID: 37591873 PMCID: PMC10435479 DOI: 10.1038/s41408-023-00899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 07/19/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
T-lymphocytes are prevalent in the tumor microenvironment of follicular lymphoma (FL). However, the phenotype of T-cells may vary, and the prevalence of certain T-cell subsets may influence tumor biology and patient survival. We therefore analyzed a cohort of 82 FL patients using CyTOF to determine whether specific T-cell phenotypes were associated with distinct tumor microenvironments and patient outcome. We identified four immune subgroups with differing T-cell phenotypes and the prevalence of certain T-cell subsets was associated with patient survival. Patients with increased T cells with early differentiation stage tended to have a significantly better survival than patients with increased T-cells of late differentiation stage. Specifically, CD57+ TFH cells, with a late-stage differentiation phenotype, were significantly more abundant in FL patients who had early disease progression and therefore correlated with an inferior survival. Single cell analysis (CITE-seq) revealed that CD57+ TFH cells exhibited a substantially different transcriptome from CD57- TFH cells with upregulation of inflammatory pathways, evidence of immune exhaustion and susceptibility to apoptosis. Taken together, our results show that different tumor microenvironments among FL patients are associated with variable T-cell phenotypes and an increased prevalence of CD57+ TFH cells is associated with poor patient survival.
Collapse
Affiliation(s)
- Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hongyan Wu
- Department of Immunology, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Jordan Krull
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Raymond M Moore
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew J Maurer
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joshua C Pritchett
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rekha Mudappathi
- Department of Quantitative Health Sciences and center for Individual Medicine, Mayo Clinic, Scottsdale, AZ, USA
- College of Health Solutions, Arizona State University, Scottsdale, AZ, USA
| | - Junwen Wang
- Department of Quantitative Health Sciences and center for Individual Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Jose C Villasboas
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Patrizia Mondello
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
12
|
Anagnostou T, Yang ZZ, Jalali S, Kim HJ, Larson DP, Tang X, Yu Y, Pritchett JC, Bisneto JV, Price-Troska TL, Mondello P, Novak AJ, Ansell SM. Characterization of immune exhaustion and suppression in the tumor microenvironment of splenic marginal zone lymphoma. Leukemia 2023; 37:1485-1498. [PMID: 37117318 DOI: 10.1038/s41375-023-01911-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
The role of the tumor microenvironment (TME) and intratumoral T cells in splenic marginal zone lymphoma (sMZL) is largely unknown. In the present study, we evaluated 36 sMZL spleen specimens by single cell analysis to gain a better understanding of the TME in sMZL. Using mass cytometry (CyTOF), we observed that the TME in sMZL is distinct from that of control non-malignant reactive spleen (rSP). We found that the number of TFH cells varied greatly in sMZL, ICOS+ TFH cells were more abundant in sMZL than rSP, and TFH cells positively correlated with increased numbers of memory B cells. Treg cell analysis revealed that TIGIT+ Treg cells are enriched in sMZL and correlate with suppression of TH17 and TH22 cells. Intratumoral CD8+ T cells were comprised of subsets of short-lived, exhausted and late-stage differentiated cells, thereby functionally impaired. We observed that T-cell exhaustion was present in sMZL and TIM-3 expression on PD-1low cells identified cells with severe immune dysfunction. Gene expression profiling by CITE-seq analysis validated this finding. Taken together, our data suggest that the TME as a whole, and T-cell population specifically, are heterogenous in sMZL and immune exhaustion is one of the major factors impairing T-cell function.
Collapse
Affiliation(s)
- Theodora Anagnostou
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Joshua C Pritchett
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Patrizia Mondello
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
13
|
Lei T, Wu G, Xu Y, Zhuang W, Lu J, Han S, Zhuang Y, Dong X, Yang H. Peripheral immune cell profiling of double-hit lymphoma by mass cytometry. BMC Cancer 2023; 23:184. [PMID: 36823603 PMCID: PMC9948356 DOI: 10.1186/s12885-023-10657-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Double-hit or Triple-hit lymphoma (DHL/THL) is a subset of high-grade B cell lymphoma harboring rearrangements of MYC and BCL2 and/or BCL6, and usually associate with aggressive profile, while current therapies tend to provide poor clinical outcomes and eventually relapsed. Further explorations of DHL at cellular and molecular levels are in demand to offer guidance for clinical activity. METHODS We collected the peripheral blood of DHL patients and diffused large B cell lymphoma (DLBCL) patients from single institute and converted them into PBMC samples. Mass cytometry was then performed to characterize these samples by 42 antibody markers with samples of healthy people as control. We divided the immune cell subtypes based on the expression profile of surface antigens, and the proportion of each cell subtype was also analyzed. By comparing the data of the DLBCL group and the healthy group, we figured out the distinguished immune cell subtypes of DHL patients according to their abundance and marker expression level. We further analyzed the heterogeneity of DHL samples by pairwise comparison based on clinical characteristics. RESULTS We found double-positive T cells (DPT) cells were in a significantly high percentage in DHL patients, whereas the ratio of double-negative T cells (DNT) was largely reduced in patients. Besides, CD38 was uniquely expressed at a high level on some naïve B cells of DHL patients, which could be a marker for the diagnosis of DHL (distinguishing from DLBCL), or even be a drug target for the treatment of DHL. In addition, we illustrated the heterogeneity of DHL patients in terms of immune cell landscape, and highlighted TP53 as a major factor that contributes to the heterogeneity of the T cells profile. CONCLUSION Our study demonstrated the distinct peripheral immune cell profile of DHL patients by contrast to DLBCL patients and healthy people, as well as the heterogeneity within the DHL group, which could provide valuable guidance for the diagnosis and treatment of DHL.
Collapse
Affiliation(s)
- Tao Lei
- grid.410726.60000 0004 1797 8419Department of Lymphoma, Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, P. R. China
| | - Gongqiang Wu
- grid.268099.c0000 0001 0348 3990Department of Hematology, Dongyang People’s Hospital, Dongyang Hospital Affiliated to Wenzhou Medical University, Dongyang, Zhejiang P. R. China
| | - Yongjin Xu
- grid.410726.60000 0004 1797 8419Department of Lymphoma, Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, P. R. China
| | - Weihao Zhuang
- grid.13402.340000 0004 1759 700XHangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Jialiang Lu
- grid.13402.340000 0004 1759 700XHangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Shuiyun Han
- grid.410726.60000 0004 1797 8419Department of Lymphoma, Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, P. R. China
| | - Yuxin Zhuang
- grid.13402.340000 0004 1759 700XHangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China
| | - Xiaowu Dong
- Hangzhou Institute of Innovative Medicine, Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China. .,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, P. R. China. .,Cancer Center, Zhejiang University, Hangzhou, P. R. China.
| | - Haiyan Yang
- Department of Lymphoma, Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Chinese Academy of Sciences, Hangzhou, P. R. China.
| |
Collapse
|
14
|
Perrett M, Edmondson C, Okosun J. Biology of follicular lymphoma: insights and windows of clinical opportunity. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:688-694. [PMID: 36485095 PMCID: PMC9820323 DOI: 10.1182/hematology.2022000361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Follicular lymphoma (FL) is a heterogeneous disease, both clinically and biologically. The biological behavior and development of FL is a culmination of complex multistep processes underpinned by genetic and nongenetic determinants. Epigenetic deregulation through recurrent genetic alterations is now a recognized major biological hallmark of FL, alongside the t(14;18) translocation. In parallel, there is a strong interplay between the lymphoma B cells and the immune microenvironment, with the microenvironment serving as a critical enabler by creating a tumor-supportive niche and modulating the immune response to favor survival of the malignant B cells. A further layer of complexity arises from the biological heterogeneity that occurs between patients and within an individual, both over the course of the disease and at different sites of disease involvement. Altogether, taking the first steps to bridge the understanding of these various biological components and how to evaluate these clinically may aid and inform future strategies, including logical therapeutic interventions, risk stratification, therapy selection, and disease monitoring.
Collapse
Affiliation(s)
- Megan Perrett
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Carina Edmondson
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
15
|
Tang X, Yang ZZ, Kim HJ, Anagnostou T, Yu Y, Wu X, Chen J, Krull JE, Wenzl K, Mondello P, Bhardwaj V, Wang J, Novak AJ, Ansell SM. Phenotype, Function, and Clinical Significance of CD26+ and CD161+Tregs in Splenic Marginal Zone Lymphoma. Clin Cancer Res 2022; 28:4322-4335. [PMID: 35686915 PMCID: PMC10443733 DOI: 10.1158/1078-0432.ccr-22-0977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/17/2022] [Accepted: 06/08/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE Regulatory T-cells (Treg) are essential to Tregs homeostasis and modulate the antitumor immune response in patients with lymphoma. However, the biology and prognostic impact of Tregs in splenic marginal zone lymphoma (SMZL) have not been studied. EXPERIMENTAL DESIGN Biopsy specimens from 24 patients with SMZL and 12 reactive spleens (rSP) from individuals without lymphoma were analyzed by using CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing), CyTOF (mass cytometry) analysis, and flow cytometry to explore the phenotype, transcriptomic profile, and clinical significance of intratumoral Tregs and their subsets. The biological characteristics and cell signaling pathways of intratumoral Treg subsets were confirmed by in vitro functional assays. RESULTS We found that Tregs are more abundant in SMZL patients' spleens than rSP, and Tregs from patients with SMZL and rSP can be separated into CD161+Treg and CD26+Treg subsets. CD161+Tregs are increased in SMZL but have dysregulated immune function. We found that CD161+Treg and CD26+Tregs have unique gene expression and phenotypic profiles and are differentially correlated with patient outcomes. Specifically, increased CD161+Tregs are significantly associated with a favorable prognosis in patients with SMZL, whereas CD26+Tregs are associated with a poor prognosis. Furthermore, activation of the IL2/STAT5 pathway contributes to the induction of CD26+Tregs and can be reversed by STAT5 inhibition. CONCLUSIONS IL2/STAT5-mediated expansion of CD26+Tregs contributes to a poor clinical outcome in SMZL and may represent a therapeutic opportunity in this disease.
Collapse
Affiliation(s)
- Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Theodora Anagnostou
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yue Yu
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Xiaosheng Wu
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jun Chen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Jordan E. Krull
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kerstin Wenzl
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Patrizia Mondello
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Vaishali Bhardwaj
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Junwen Wang
- Department of Quantitative Health Sciences and Center for Individualized Medicine, Mayo Clinic, Scottsdale, Arizona
| | - Anne J. Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Stephen M. Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
16
|
Liu L, Yu X, Li Z, He X, Zha J, Lin Z, Hong Y, Zheng H, Lai Q, Ding K, Jia X, Fu G, Yu H, Yang H, Li Z, Young KH, Xu B. Revealing the evolution of the tumor immune microenvironment in follicular lymphoma patients progressing within 24 months using single-cell imaging mass cytometry. J Hematol Oncol 2022; 15:115. [PMID: 35996180 PMCID: PMC9396877 DOI: 10.1186/s13045-022-01326-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/04/2022] [Indexed: 11/11/2022] Open
Abstract
Background Patients with follicular lymphoma (FL) who experience disease progression within 24 months (POD24) have inferior outcomes. The tumor immune microenvironment (TIME) plays a crucial role in pathogenesis and progression of follicular lymphoma (FL). However, TIME evolution during progression of disease within 24 months (POD24) is elusive. Methods Spatially resolved and single-cell image mass cytometry with a panel of 36 metal-tagged antibodies was used to quantitatively analyze the TIME structure in 13 paired FLs at diagnosis and POD24. Results Follicles and peri-follicular regions were well dissected in structure. Peri-follicular regions represented a barrier for immune infiltration into the follicles. More FL-cells in the peri-follicular regions suffered CD8+T cells attacks under simultaneous protection of regulatory T cells (Tregs) and/or macrophages compared with that in the follicles irrespective of POD24. During POD24, increased CD163− macrophages with PD-1 ligand upregulation and decreased CD8+T cells with upregulated LAG-3 expression around FL-cells were observed in the follicles. Spatial analyses demonstrated that FL-cells interacted more intimately with macrophages than with Tregs and less with cytotoxic T cells in both peri-follicular regions and follicles during POD24. In comparison, macrophages also cooperated more frequently with Tregs to simultaneously hijack FL-cells, creating an enhanced immunosuppressive environment in both peri-follicular and follicular regions during POD24. Conclusions Peri-follicular regions function as a barrier by recruiting both CD8+T cells and immunosuppressive cells, protecting follicular FL-cells from immune attack at diagnosis or POD24. FL-cells reside in a more immune-compromised microenvironment and evade immune cell attacks during POD24. Novel immunotherapeutic approaches harnessing LAG-3, macrophages, and Tregs will be empowered to overcome poor outcomes in patients with FL POD24. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01326-z.
Collapse
Affiliation(s)
- Long Liu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Xingxing Yu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Zhifeng Li
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Xiaohua He
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, People's Republic of China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Zhijuan Lin
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Yan Hong
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Huijian Zheng
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Qian Lai
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China.,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China
| | - Kaiyang Ding
- Department of Hematology, The First Affiliated Hospital of USTC Anhui Provincial Hospital, Hefei, 230001, People's Republic of China
| | - Xian Jia
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Haifeng Yu
- Department of Lymphoma, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310012, People's Republic of China.,Department of Lymphoma, Institute of Cancer, and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, 310012, People's Republic of China
| | - Haiyan Yang
- Department of Lymphoma, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310012, People's Republic of China.,Department of Lymphoma, Institute of Cancer, and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, 310012, People's Republic of China
| | - Zhiming Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, People's Republic of China.
| | - Ken H Young
- Hematopathology Division, Department of Pathology, Duke University Medical Center, Durham, NC, 27710, USA. .,Duke University Cancer Institute, Durham, NC, USA.
| | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, 361003, People's Republic of China. .,Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, 361003, People's Republic of China.
| |
Collapse
|
17
|
Gršković P, Hančić S, Dotlić S, Matulić M, Ostojić Kolonić S, Gašparov S, Dominis M, Korać P. CD4+/CD57+/CD69+ T lymphocytes and CD14+ dendritic cells accumulate in advanced follicular lymphoma. Immunobiology 2022; 227:152257. [PMID: 35964344 DOI: 10.1016/j.imbio.2022.152257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/11/2022] [Accepted: 08/05/2022] [Indexed: 11/05/2022]
Abstract
Follicular lymphoma is the second most frequent non-Hodgkin's lymphoma, accounting for around 20 % of all lymphomas in Western countries. Initially, it behaves indolently, but in time becomes more aggressive and less susceptible to chemotherapy. Multiple features correlate with the survival of the patients and the progression of the disease, such as therapy with rituximab, tumour microenvironment and the intrafollicular proliferation index. Our research was focused on the association of specific components of tumour microenvironment and the tumour behaviour. The presence and the relative percentage of T lymphocytes, follicular dendritic cells, dendritic cells and macrophages was detected by immunohistochemical staining of the antigens specific for certain cell populations. Our results show that T lymphocytes and dendritic cells affect tumour growth, possibly through interactions with tumour cells. Higher patients' ECOG score and the outcome of the disease are associated with the presence of CD14+ dendritic cells in tumour tissue, while the worse overall survival of patients is associated with the increased number of activated helper T lymphocytes that express marker of exhaustion CD57. Taken together, our results suggest that the efficiency of the immune response against follicular lymphoma depends on more than one type of immune cells. Also, we found that the phenotype of these cells, rather than just their number, affects the tumour behaviour and in consequence survival of the patients.
Collapse
Affiliation(s)
- Paula Gršković
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia.
| | - Suzana Hančić
- Institute of Clinical Pathology and Cytology, Merkur University Hospital, Zagreb, Croatia
| | - Snježana Dotlić
- Department of Pathology and Cytology, University Hospital Centre Zagreb, Zagreb, Croatia; Department of Pathology, Medical School Zagreb, University of Zagreb, Zagreb, Croatia
| | - Maja Matulić
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia.
| | - Slobodanka Ostojić Kolonić
- Department of Internal Medicine, Medical School Zagreb, University of Zagreb, Zagreb, Croatia; Division of Haematology, Department of Internal Medicine, Merkur University Hospital, Zagreb, Croatia.
| | - Slavko Gašparov
- Institute of Clinical Pathology and Cytology, Merkur University Hospital, Zagreb, Croatia; Department of Pathology, Medical School Zagreb, University of Zagreb, Zagreb, Croatia
| | - Mara Dominis
- Institute of Clinical Pathology and Cytology, Merkur University Hospital, Zagreb, Croatia; Department of Pathology, Medical School Zagreb, University of Zagreb, Zagreb, Croatia
| | - Petra Korać
- Division of Molecular Biology, Department of Biology, Faculty of Science, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
18
|
LINC00892 Is an lncRNA Induced by T Cell Activation and Expressed by Follicular Lymphoma-Resident T Helper Cells. Noncoding RNA 2022; 8:ncrna8030040. [PMID: 35736637 PMCID: PMC9228450 DOI: 10.3390/ncrna8030040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
Successful immunotherapy in both solid tumors and in hematological malignancies relies on the ability of T lymphocytes to infiltrate the cancer tissue and mount an immune response against the tumor. Biomarkers able to discern the amount and the types of T lymphocytes infiltrating a given tumor therefore have high diagnostic and prognostic value. Given that lncRNAs are known to have a highly cell-type-specific expression pattern, we searched for lncRNAs specifically expressed by activated T cells and at the same time in a kind of lymphoma, follicular lymphoma, where the microenvironment is known to play a critical role in the regulation of antitumor immunity. We focused on a non-coding transcript, annotated as LINC00892, which reaches extremely high expression levels following cell activation in Jurkat cells. Interestingly LINC00892 has an expression pattern resembling that of genes involved in T cell memory. Accordingly, LINC00892 is mostly expressed by the effector memory and helper CD4+ T cell sub-types but not by naïve T cells. In situ analyses of LINC00892 expression in normal lymph nodes and in follicular lymphoma biopsies show that its expression is limited to CD4+ PD1hi T cells, with a subcellular localization within the germinal center matching that of follicular helper T cells. Our analysis therefore suggests that the previously uncharacterized lncRNA LINC00892 could be a useful biomarker for the detection of CD4+ memory T cells in both normal and tumor tissues.
Collapse
|
19
|
Seal S, Vu T, Ghosh T, Wrobel J, Ghosh D. DenVar: density-based variation analysis of multiplex imaging data. BIOINFORMATICS ADVANCES 2022; 2:vbac039. [PMID: 36699398 PMCID: PMC9710661 DOI: 10.1093/bioadv/vbac039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/17/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023]
Abstract
Summary Multiplex imaging platforms have become popular for studying complex single-cell biology in the tumor microenvironment (TME) of cancer subjects. Studying the intensity of the proteins that regulate important cell-functions becomes extremely crucial for subject-specific assessment of risks. The conventional approach requires selection of two thresholds, one to define the cells of the TME as positive or negative for a particular protein, and the other to classify the subjects based on the proportion of the positive cells. We present a threshold-free approach in which distance between a pair of subjects is computed based on the probability density of the protein in their TMEs. The distance matrix can either be used to classify the subjects into meaningful groups or can directly be used in a kernel machine regression framework for testing association with clinical outcomes. The method gets rid of the subjectivity bias of the thresholding-based approach, enabling easier but interpretable analysis. We analyze a lung cancer dataset, finding the difference in the density of protein HLA-DR to be significantly associated with the overall survival and a triple-negative breast cancer dataset, analyzing the effects of multiple proteins on survival and recurrence. The reliability of our method is demonstrated through extensive simulation studies. Availability and implementation The associated R package can be found here, https://github.com/sealx017/DenVar. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- Souvik Seal
- Department of Biostatistics and Informatics, University of Colorado CU Anschutz Medical Campus, Aurora, CO, USA,To whom correspondence should be addressed.
| | - Thao Vu
- Department of Biostatistics and Informatics, University of Colorado CU Anschutz Medical Campus, Aurora, CO, USA
| | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, University of Colorado CU Anschutz Medical Campus, Aurora, CO, USA
| | - Julia Wrobel
- Department of Biostatistics and Informatics, University of Colorado CU Anschutz Medical Campus, Aurora, CO, USA
| | - Debashis Ghosh
- Department of Biostatistics and Informatics, University of Colorado CU Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
20
|
Bae J, Kim JE, Perumalsamy H, Park S, Kim Y, Jun DW, Yoon TH. Mass Cytometry Study on Hepatic Fibrosis and Its Drug-Induced Recovery Using Mouse Peripheral Blood Mononuclear Cells. Front Immunol 2022; 13:814030. [PMID: 35222390 PMCID: PMC8863676 DOI: 10.3389/fimmu.2022.814030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/03/2022] [Indexed: 01/10/2023] Open
Abstract
The number of patients with liver diseases has increased significantly with the progress of global industrialization. Hepatic fibrosis, one of the most common liver diseases diagnosed in many developed countries, occurs in response to chronic liver injury and is primarily driven by the development of inflammation. Earlier immunological studies have been focused on the importance of the innate immune response in the pathophysiology of steatohepatitis and fibrosis, but recently, it has also been reported that adaptive immunity, particularly B cells, plays an essential role in hepatic inflammation and fibrosis. However, despite recent data showing the importance of adaptive immunity, relatively little is known about the role of B cells in the pathogenesis of steatohepatitis fibrosis. In this study, a single-cell-based, high-dimensional mass cytometric investigation of the peripheral blood mononuclear cells collected from mice belonging to three groups [normal chow (NC), thioacetamide (TAA), and 11beta-HSD inhibitor drug] was conducted to further understand the pathogenesis of liver fibrosis through reliable noninvasive biomarkers. Firstly, major immune cell types and their population changes were qualitatively analyzed using UMAP dimensionality reduction and two-dimensional visualization technique combined with a conventional manual gating strategy. The population of B cells displayed a twofold increase in the TAA group compared to that in the NC group, which was recovered slightly after treatment with the 11beta-HSD inhibitor drug. In contrast, the populations of NK cells, effector CD4+ T cells, and memory CD8+ T cells were significantly reduced in the TAA group compared with those in the NC group. Further identification and quantification of the major immune cell types and their subsets were conducted based on automated clustering approaches [PhenoGraph (PG) and FlowSOM]. The B-cell subset corresponding to PhenoGraph cluster PG#2 (CD62LhighCD44highLy6chigh B cells) and PG#3 (CD62LhighCD44highLy6clow B cell) appears to play a major role in both the development of hepatic fibrosis and recovery via treatment, whereas PG#1 (CD62LlowCD44highLy6clow B cell) seems to play a dominant role in the development of hepatic fibrosis. These findings provide insights into the roles of cellular subsets of B cells during the progression of, and recovery from, hepatic fibrosis.
Collapse
Affiliation(s)
- Jiwon Bae
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - Ji Eun Kim
- Department of Internal Medicine, Hanyang University Hospital, Seoul, South Korea
| | - Haribalan Perumalsamy
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, South Korea
| | - Sehee Park
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, South Korea
| | - Yun Kim
- Hanyang Medicine-Engineering-Bio Collaborative & Comprehensive Center for Drug Development, Hanyang University, Seoul, South Korea.,Department of Clinical Pharmacology and Therapeutics, Hanyang University Hospital, Seoul, South Korea
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University Hospital, Seoul, South Korea.,Hanyang Medicine-Engineering-Bio Collaborative & Comprehensive Center for Drug Development, Hanyang University, Seoul, South Korea.,Department of Medical and Digital Engineering, College of Engineering, Hanyang University, Seoul, South Korea
| | - Tae Hyun Yoon
- Department of Chemistry, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Convergence of Basic Science, Hanyang University, Seoul, South Korea.,Institute of Next Generation Material Design, Hanyang University, Seoul, South Korea.,Yoon Idea Lab. Co. Ltd, Seoul, South Korea
| |
Collapse
|
21
|
Husby S, Jørgensen GØ, Favero F, Jespersen JS, Rodriguez-Gonzalez FG, Nielsen C, Sorensen B, Ebbesen LH, Bæch J, Haastrup EK, Josefsson P, Thorsgaard M, Brown P, El-Galaly TC, Larsen TS, Weischenfeldt J, Grønbæk K. Level of unique T cell clonotypes is associated with clonal hematopoiesis and survival in patients with lymphoma undergoing ASCT. Bone Marrow Transplant 2022; 57:674-677. [PMID: 35124694 DOI: 10.1038/s41409-022-01580-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/20/2021] [Accepted: 01/12/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Simon Husby
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark.
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Gustav Ø Jørgensen
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Francesco Favero
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Jakob Schmidt Jespersen
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Francisco G Rodriguez-Gonzalez
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Christian Nielsen
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, Odense, Denmark
| | - Betina Sorensen
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Lene H Ebbesen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - John Bæch
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Eva K Haastrup
- Department of Clinical Immunology, Rigshospitalet, Copenhagen, Denmark
| | - Pär Josefsson
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | | | - Peter Brown
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
| | - Tarec C El-Galaly
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Thomas Stauffer Larsen
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, Odense, Denmark
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Joachim Weischenfeldt
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, DanStem, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Canine memory T-cell subsets in health and disease. Vet Immunol Immunopathol 2022; 246:110401. [PMID: 35255296 DOI: 10.1016/j.vetimm.2022.110401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 11/22/2022]
Abstract
A more complete understanding of canine T-lymphocyte immunity is necessary for improving diagnostic and therapeutic approaches to canine diseases, developing cell-based canine immunotherapeutics, and evaluating dogs as large mammal models for comparative immunology research. The aim of this study was to utilize CD45RA (indicating antigen inexperience) and CD62L (indicating lymph node homing capability), to quantify canine memory T-cell subsets in healthy dogs and dogs with various diseases. Peripheral blood mononuclear cells (PBMCs) were prospectively collected from dogs belonging to one of four groups:dermatologic inflammation (n = 9), solid tumors (n = 9), lymphoma (n = 9), and age-/weight-matched healthy control dogs (n = 15). Dogs receiving prednisone or any other immunomodulating medication within two weeks were excluded. Flow cytometry was performed and T-cell subsets were defined as CD4+ or CD8+, and naïve (TN), central memory (CM), effector memory (EM), or terminal effector memory re-expressing CD45RA (TEMRA). T-cell subset proportions were compared between each disease group and their healthy age-/weight-matched controls using a Mann-Whitney test. Significantly increased %CD8+ TN (P = 0.036) and decreased %CD8+ TEMRA (P = 0.045) were detected in dogs with dermatologic inflammation compared to healthy controls. Furthermore, %CD4+ TN positively correlated with Canine Atopic Dermatitis Extent and Severity Index (CADESI) score within the inflammation group (ρ = 0.817, P = 0.011). No significant differences between either cancer group and their healthy controls were detected. Taken together, these data indicate that dermatologic inflammation can alter proportions of peripheral blood T-cell subsets, possibly due to the migration of antigen-specific T-cells into tissues. Furthermore, these findings support the utility of CD45RA and CD62L in characterizing clinical canine immune responses.
Collapse
|
23
|
Astle JM, Huang H. Mass Cytometry in Hematologic Malignancies: Research Highlights and Potential Clinical Applications. Front Oncol 2021; 11:704464. [PMID: 34858804 PMCID: PMC8630615 DOI: 10.3389/fonc.2021.704464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Recent advances in global gene sequencing technologies and the effect they have had on disease diagnosis, therapy, and research have fueled interest in technologies capable of more broadly profiling not only genes but proteins, metabolites, cells, and almost any other component of biological systems. Mass cytometry is one such technology, which enables simultaneous characterization of over 40 parameters per cell, significantly more than can be achieved by even the most state-of-the-art flow cytometers. This mini-review will focus on how mass cytometry has been utilized to help advance the field of neoplastic hematology. Common themes among published studies include better defining lineage sub-populations, improved characterization of tumor microenvironments, and profiling intracellular signaling across multiple pathways simultaneously in various cell types. Reviewed studies highlight potential applications for disease diagnosis, prognostication, response to therapy, measurable residual disease analysis, and identifying new therapies.
Collapse
Affiliation(s)
- John M Astle
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Huiya Huang
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
24
|
Visram A, Kourelis TV. Aging-associated immune system changes in multiple myeloma: The dark side of the moon. Cancer Treat Res Commun 2021; 29:100494. [PMID: 34837796 DOI: 10.1016/j.ctarc.2021.100494] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 11/18/2021] [Indexed: 02/08/2023]
Abstract
Multiple myeloma (MM) is a disease of the elderly. Changes that occur in the immune system with aging, also known as immunosenescence, have been associated with decreased tumor immunosurveillance and are thought to contribute to the development of MM and other cancers in the elderly. Once MM establishes itself in the bone marrow, immunosenescence related changes have been observed in the immune tumor microenvironment (iTME) and are driven by the malignant cells. The efficacy of novel immunotherapies used to treat MM has been blunted by detrimental iTME changes that occur at later disease stages and are, to some extent, driven by prior therapies. In this review, we discuss general changes that occur in the immune system with aging as well as our current knowledge of immunosenescence in MM. We discuss the differences and overlap between T cell senescence and exhaustion as well as potential methods to prevent or reverse immunosenescence. We focus predominantly on T cell immunosenescence which has been better evaluated in this disease and is more pertinent to novel MM immunotherapies. Our lack of understanding of the drivers of immunosenescence at each stage of the disease, from precursor stages to heavily pretreated MM, represents a major barrier to improving the efficacy of novel and existing therapies.
Collapse
Affiliation(s)
- Alissa Visram
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN United States; Department of Medicine, Division of Hematology, University of Ottawa, Ottawa Hospital Research Institute, Ontario, Canada
| | - Taxiarchis V Kourelis
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN United States.
| |
Collapse
|
25
|
Kumar E, Pickard L, Okosun J. Pathogenesis of follicular lymphoma: genetics to the microenvironment to clinical translation. Br J Haematol 2021; 194:810-821. [PMID: 33694181 DOI: 10.1111/bjh.17383] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/08/2021] [Indexed: 01/10/2023]
Abstract
Follicular lymphoma (FL) represents a heterogeneous disease both clinically and biologically. The pathognomonic t(14;18) translocation can no longer be thought of as the primary genetic driver, with increasing recognition of the biological relevance of recurrent genetic alterations in epigenetic regulators that now feature as a pivotal hallmark of this lymphoma subtype. Furthermore, sequencing studies have provided a near complete catalogue of additional genetic aberrations. Longitudinal and spatial genetic studies add an additional layer to the biological heterogeneity, providing preliminary molecular insights into high-risk phenotypes such as early progressors and transformation, and also supporting evidence for the existence of persisting re-populating cells that act as lymphoma reservoirs and harbingers for FL recurrence. Simultaneously, understanding of the tumour microenvironmental cues promoting lymphomagenesis and disease progression continue to broaden. More recently, studies are beginning to unravel the convergence and co-operation between the genetics, epigenetics and microenvironment. There is a pressing need to marry biology with therapeutics, especially with the burgeoning treatment landscape in FL, to aid in optimising patient selection and guiding the 'right drug to the right patient'.
Collapse
Affiliation(s)
- Emil Kumar
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Lucy Pickard
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
26
|
Mondello P, Fama A, Larson MC, Feldman AL, Villasboas JC, Yang ZZ, Galkin I, Svelolkin V, Postovalova E, Bagaev A, Ovcharov P, Varlamova A, Huet S, Tesson B, McGrath KR, Slager S, Link BK, Syrbu S, Novak AJ, Habermann TM, Witzig TE, Nowakowski GS, Salles G, Cerhan JR, Ansell SM. Lack of intrafollicular memory CD4 + T cells is predictive of early clinical failure in newly diagnosed follicular lymphoma. Blood Cancer J 2021; 11:130. [PMID: 34267181 PMCID: PMC8282842 DOI: 10.1038/s41408-021-00521-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/24/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Despite a characteristic indolent course, a substantial subset of follicular lymphoma (FL) patients has an early relapse with a poor outcome. Cells in the microenvironment may be a key contributor to treatment failure. We used a discovery and validation study design to identify microenvironmental determinants of early failure and then integrated these results into the FLIPI. In total, 496 newly diagnosed FL grade 1-3 A patients who were prospectively enrolled into the MER cohort from 2002 to 2012 were evaluated. Tissue microarrays were stained for CD4, CD8, FOXP3, CD32b, CD14, CD68, CD70, SIRP-α, TIM3, PD-1, and PD-L1. Early failure was defined as failing to achieve event-free survival at 24 months (EFS24) in immunochemotherapy-treated patients and EFS12 in all others. CyTOF and CODEX analysis were performed to characterize intratumoral immunophenotypes. Lack of intrafollicular CD4 expression was the only predictor of early failure that replicated with a pooled OR 2.37 (95%CI 1.48-3.79). We next developed a bio-clinical risk model (BioFLIPI), where lack of CD4 intrafollicular expression moved patients up one FLIPI risk group, adding a new fourth high-risk group. Compared with BioFLIPI score of 1, patients with a score of 2 (OR 2.17; 95% CI 1.08-4.69), 3 (OR 3.53; 95% CI 1.78-7.54), and 4 (OR 8.92; 95% CI 4.00-21.1) had increasing risk of early failure. The favorable intrafollicular CD4 T cells were identified as activated central memory T cells, whose prognostic value was independent from genetic features. In conclusion, lack of intrafollicular CD4 expression predicts early failure in FL and combined with FLIPI improves identification of high-risk patients; however, independent validation is warranted.
Collapse
Affiliation(s)
- Patrizia Mondello
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Angelo Fama
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
- Hematology Unit, Arcispedale Santa Maria Nuova, Azienda Unità Sanitaria Locale- IRCCS, Reggio Emilia, Italy
| | - Melissa C Larson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Jose C Villasboas
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | - Sarah Huet
- Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, laboratoire d'hématologie, Pierre-Bénite, France
- Université Claude Bernard Lyon I, Lyon, France
| | | | - Kaitlyn R McGrath
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Susan Slager
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Brian K Link
- Division of Hematology, Oncology and Bone Marrow Transplantation, University of Iowa, Iowa City, IA, USA
| | - Sergei Syrbu
- Department of Pathology, University of Iowa, Iowa City, IA, USA
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas M Habermann
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Thomas E Witzig
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Gilles Salles
- Université Claude Bernard Lyon I, Lyon, France
- Hospices Civils de Lyon, Centre Hospitalier Lyon Sud, service d'Hématologie, Pierre-Bénite, France
| | - James R Cerhan
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
27
|
Yao H, Zhao H, Pan X, Zhao X, Feng J, Yang C, Zhang S, Zhang X. Discriminating Leukemia Cellular Heterogeneity and Screening Metabolite Biomarker Candidates using Label-Free Mass Cytometry. Anal Chem 2021; 93:10282-10291. [PMID: 34259005 DOI: 10.1021/acs.analchem.1c01746] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Discriminating various leukocyte subsets with specific functions is critical due to their important roles in the development of many diseases. Here, we proposed a general strategy to unravel leukocytes heterogeneity and screen differentiated metabolites as biomarker candidates for leukocyte subtypes using the label-free mass cytometry (CyESI-MS) combined with a homemade data processing workflow. Taking leukemia cells as an example, metabolic fingerprints of single leukemia cells were obtained from 472 HL-60, 416 THP-1, 313 U937, 356 Jurkat, and 366 Ramos cells, with throughput up to 40 cells/min. Five leukemia subtypes were clearly distinguished by unsupervised learning t-SNE analysis of the single-cell metabolic fingerprints. Cell discrimination in the mixed leukemia cell samples was also realized by supervised learning of the single-cell metabolic fingerprints with high recovery and good repetition (98.31 ± 0.24%, -102.35 ± 4.82%). Statistical analysis and metabolite assignment were carried out to screen characteristic metabolites for discrimination and 36 metabolites with significant differences were annotated. Then, differentiated metabolites for pairwise discrimination of five leukemia subtypes were further selected as biomarker candidates. Furthermore, discriminating cultured leukemia cells from human normal leukocytes, separated from fresh human peripheral blood, was performed based on single-cell metabolic fingerprints as well as the proposed biomarker candidates, unveiling the potential of this strategy in clinical research. This work makes efforts to realize high-throughput single-leukocyte metabolic analysis and metabolite-based discrimination of leukocytes. It is expected to be a powerful means for the clinical molecular diagnosis of hematological diseases.
Collapse
Affiliation(s)
- Huan Yao
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Hansen Zhao
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Xingyu Pan
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Xu Zhao
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Jiaxin Feng
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Chengdui Yang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Sichun Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Xinrong Zhang
- Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
28
|
Pritchett JC, Yang ZZ, Kim HJ, Villasboas JC, Tang X, Jalali S, Cerhan JR, Feldman AL, Ansell SM. High-dimensional and single-cell transcriptome analysis of the tumor microenvironment in angioimmunoblastic T cell lymphoma (AITL). Leukemia 2021; 36:165-176. [PMID: 34230608 DOI: 10.1038/s41375-021-01321-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/20/2021] [Accepted: 06/05/2021] [Indexed: 02/08/2023]
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is an aggressive lymphoid malignancy associated with a poor clinical prognosis. The AITL tumor microenvironment (TME) is unique, featuring a minority population of malignant CD4+ T follicular helper (TFH) cells inter-mixed with a diverse infiltrate of multi-lineage immune cells. While much of the understanding of AITL biology to date has focused on characteristics of the malignant clone, less is known about the many non-malignant populations that comprise the TME. Recently, mutational consistencies have been identified between malignant cells and non-malignant B cells within the AITL TME. As a result, a significant role for non-malignant populations in AITL biology has been increasingly hypothesized. In this study, we have utilized mass cytometry and single-cell transcriptome analysis to identify several expanded populations within the AITL TME. Notably, we find that B cells within the AITL TME feature decreased expression of key markers including CD73 and CXCR5. Furthermore, we describe the expansion of distinct CD8+ T cell populations that feature an exhausted phenotype and an underlying expression profile indicative of dysfunction, impaired cytotoxicity, and upregulation of the chemokines XCL2 and XCL1.
Collapse
Affiliation(s)
| | - Zhi-Zhang Yang
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Hyo Jin Kim
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Xinyi Tang
- Department of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - James R Cerhan
- Department of Health Sciences Research and Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Andrew L Feldman
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
29
|
Wu H, Tang X, Kim HJ, Jalali S, Pritchett JC, Villasboas JC, Novak AJ, Yang ZZ, Ansell SM. Expression of KLRG1 and CD127 defines distinct CD8 + subsets that differentially impact patient outcome in follicular lymphoma. J Immunother Cancer 2021; 9:jitc-2021-002662. [PMID: 34226281 PMCID: PMC8258669 DOI: 10.1136/jitc-2021-002662] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 11/15/2022] Open
Abstract
Background CD8+ T-lymphocyte subsets defined by killer lectin-like receptor G1 (KLRG1) and CD127 expression have been reported to have an important role in infection, but their role in the setting of lymphoid malignancies, specifically follicular lymphoma (FL), has not been studied. Methods To characterize the phenotype of KLRG1/CD127-defined CD8+ subsets, surface and intracellular markers were measured by flow cytometry and Cytometry by time of flight (CyTOF), and the transcriptional profile of these cells was determined by CITE-Seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing). The functional capacity of each subset was determined, as was their impact on overall survival (OS) and event-free survival (EFS) of patients with FL. Results We found that intratumoral CD8+ cells in FL are skewed toward effector cell subsets, particularly KLRG+CD127- and KLRG1-CD127- cells over memory cell subsets, such as KLRG1-CD127+ and KLRG1+CD127+ cells. While effector subsets exhibited increased capacity to produce cytokines/granules when compared with memory subsets, their proliferative capacity and viability were found to be substantially inferior. Clinically, a skewed distribution of intratumoral CD8+ T cells favoring effector subtypes was associated with an inferior outcome in patients with FL. Increased numbers of CD127+KLRG1- and CD127+KLRG1+ were significantly associated with a favorable OS and EFS, while CD127-KLRG1- correlated with a poor EFS and OS in patients with FL. Furthermore, we demonstrated that interleukin (IL)-15 promotes CD127-KLRG1+ cell development in the presence of dendritic cells via a phosphoinositide 3-kinase (PI3K)-dependent mechanism, and treatment of CD8+ T cells with a PI3K inhibitor downregulated the transcription factors responsible for CD127-KLRG1+ differentiation. Conclusions Taken together, these results reveal not only a biological and prognostic role for KLRG1/CD127-defined CD8+ subsets in FL but also a potential role for PI3K inhibitors to manipulate the differentiation of CD8+ T cells, thereby promoting a more effective antitumor immune response.
Collapse
Affiliation(s)
- Hongyan Wu
- Department of Immunology, Medical College, China Three Gorges University, Yichang, Hubei, People's Republic of China
| | - Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joshua C Pritchett
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jose C Villasboas
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
30
|
Hanel W, Epperla N. Evolving therapeutic landscape in follicular lymphoma: a look at emerging and investigational therapies. J Hematol Oncol 2021; 14:104. [PMID: 34193230 PMCID: PMC8247091 DOI: 10.1186/s13045-021-01113-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/04/2021] [Indexed: 02/08/2023] Open
Abstract
Follicular Lymphoma (FL) is the most common subtype of indolent B cell non-Hodgkin lymphoma. The clinical course can be very heterogeneous with some patients being safely observed over many years without ever requiring treatment to other patients having more rapidly progressive disease requiring multiple lines of treatment for disease control. Front-line treatment of advanced FL has historically consisted of chemoimmunotherapy but has extended to immunomodulatory agents such as lenalidomide. In the relapsed setting, several exciting therapies that target the underlying biology and immune microenvironment have emerged, most notable among them include targeted therapies such as phosphoinositide-3 kinase and Enhancer of Zeste 2 Polycomb Repressive Complex 2 inhibitors and cellular therapies including chimeric antigen receptor T cells and bispecific T cell engagers. There are several combination therapies currently in clinical trials that appear promising. These therapies will likely reshape the treatment approach for patients with relapsed and refractory FL in the coming years. In this article, we provide a comprehensive review of the emerging and investigational therapies in FL and discuss how these agents will impact the therapeutic landscape in FL.
Collapse
Affiliation(s)
- Walter Hanel
- Division of Hematology, Department of Medicine, The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 10th Ave, Columbus, OH 43210 USA
| | - Narendranath Epperla
- Division of Hematology, Department of Medicine, The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 10th Ave, Columbus, OH 43210 USA
- The Ohio State University Comprehensive Cancer Center, 1110E Lincoln Tower, 1800 Cannon Drive, Columbus, OH 43210 USA
| |
Collapse
|
31
|
Milpied P, Gandhi AK, Cartron G, Pasqualucci L, Tarte K, Nadel B, Roulland S. Follicular lymphoma dynamics. Adv Immunol 2021; 150:43-103. [PMID: 34176559 DOI: 10.1016/bs.ai.2021.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Follicular lymphoma (FL) is an indolent yet challenging disease. Despite a generally favorable response to immunochemotherapy regimens, a fraction of patients does not respond or relapses early with unfavorable prognosis. For the vast majority of those who initially respond, relapses will repeatedly occur with increasing refractoriness to available treatments. Addressing the clinical challenges in FL warrants deep understanding of the nature of treatment-resistant FL cells seeding relapses, and of the biological basis of early disease progression. Great progress has been made in the last decade in the description and interrogation of the (epi)genomic landscape of FL cells, of their major dependency to the tumor microenvironment (TME), and of the stepwise lymphomagenesis process, from healthy to subclinical disease and to overt FL. A new picture is emerging, in which an ever-evolving tumor-TME duo sparks a complex and multilayered clonal and functional heterogeneity, blurring the discovery of prognostic biomarkers, patient stratification and reliable designs of risk-adapted treatments. Novel technological approaches allowing to decipher both tumor and TME heterogeneity at the single-cell level are beginning to unravel unsuspected cell dynamics and plasticity of FL cells. The upcoming drawing of a comprehensive functional picture of FL within its ecosystem holds great promise to address the unmet medical needs of this complex lymphoma.
Collapse
Affiliation(s)
- Pierre Milpied
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Anita K Gandhi
- Translational Medicine, Bristol Myers Squibb, Summit, NJ, United States
| | - Guillaume Cartron
- Department of Hematology, Centre Hospitalier Universitaire Montpellier, UMR-CNRS 5535, Montpellier, France
| | - Laura Pasqualucci
- Pathology and Cell Biology, Institute for Cancer Genetics, Columbia University, New York City, NY, United States
| | - Karin Tarte
- INSERM U1236, Univ Rennes, EFS Bretagne, CHU Rennes, Rennes, France
| | - Bertrand Nadel
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France.
| | | |
Collapse
|
32
|
Lenalidomide triggers T-cell effector functions in vivo in patients with follicular lymphoma. Blood Adv 2021; 5:2063-2074. [PMID: 33877296 DOI: 10.1182/bloodadvances.2020003774] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/08/2021] [Indexed: 02/08/2023] Open
Abstract
The immunomodulatory drug lenalidomide is used in patients with follicular lymphoma (FL) with the aim of stimulating T-cell antitumor immune response. However, little is known about the effects of lenalidomide on T-cell biology in vivo in patients with FL. We thus undertook an extensive longitudinal immunologic study, including phenotypic, transcriptomic, and functional analyses, on 44 first-line and 27 relapsed/refractory patients enrolled in the GALEN trial (Obinutuzumab Combined With Lenalidomide for Relapsed or Refractory Follicular B-Cell Lymphoma) to test the efficacy of lenalidomide and obinutuzumab combination in patients with FL. Lenalidomide rapidly and transiently induced an activated T-cell phenotype, including HLA-DR, Tim-3, CD137, and programmed cell death protein 1 (PD-1) upregulation. Furthermore, sequential RNA-sequencing of sorted PD-1+ and PD-1- T-cell subsets revealed that lenalidomide triggered a strong enrichment for several gene signatures related to effector memory T-cell features, including proliferation, antigen receptor signaling, and immune synapse restoration; all were validated at the phenotypic level and with ex vivo functional assays. Correlative analyses pinpointed a negative clinical impact of high effector T-cell and regulatory T-cell percentages before and during treatment. Our findings bring new insight in lenalidomide mechanisms of action at work in vivo and will fuel a new rationale for the design of combination therapies.
Collapse
|
33
|
The Tumor Microenvironment in Follicular Lymphoma: Its Pro-Malignancy Role with Therapeutic Potential. Int J Mol Sci 2021; 22:ijms22105352. [PMID: 34069564 PMCID: PMC8160856 DOI: 10.3390/ijms22105352] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
In the follicular lymphoma (FL) microenvironment, CXCR5+ICOS+PD1+BCL6+ follicular helper T (Tfh) cells, which closely correlate with FL B cells in neoplastic follicles, play a major role in supporting FL. Interleukin-4 secreted by Tfh cells triggers the upregulation of the lymphocyte chemoattractant CXCL12 in stromal cell precursors, in particular by fibroblastic reticular cells (FRCs). In turn, mesenchymal stem cells (MSCs) can be committed to FRC differentiation in the bone marrow and lymph nodes involved by FL. Noteworthy, MSCs can promote the differentiation of Tfh cells into highly immunosuppressive T-follicular regulatory cells. The tumor suppressor HVEM is highly mutated in FL cells, and its deficiency increases Tfh cell frequency. In contrast, PI3Kδ inhibition impedes the recruitment of Tfh/regulatory T cells and impairs the proliferation of follicular dendritic cells (FDCs) and FDC-induced angiogenesis. Since TIGIT ligands are expressed by FDCs, the immune checkpoint receptor TIGIT plays an important role in tumor-infiltrating T cells. Thus, TIGIT blockade might invigorate cytotoxic T cells in the FL microenvironment. Given their potential to simultaneously reduce the neoplastic B cells, Tfh, and TFR cells could also reinforce the effects of the cytotoxic T cells. This combinatory strategy should be explored as a treatment option to tackle FL.
Collapse
|
34
|
Zha H, Jiang Y, Wang X, Shang J, Wang N, Yu L, Zhao W, Li Z, An J, Zhang X, Chen H, Zhu B, Li Z. Non-canonical PD-1 signaling in cancer and its potential implications in clinic. J Immunother Cancer 2021; 9:jitc-2020-001230. [PMID: 33593825 PMCID: PMC7888367 DOI: 10.1136/jitc-2020-001230] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Programmed cell death 1 (PD-1)-based immunotherapy has revolutionized the treatment of various cancers. However, only a certain group of patients benefit from PD-1 blockade therapy and many patients succumb to hyperprogressive disease. Although, CD8 T cells and conventional T cells are generally considered to be the primary source of PD-1 in cancer, accumulating evidence suggests that other distinct cell types, including B cells, regulatory T cells, natural killer cells, dendritic cells, tumor-associated macrophages and cancer cells, also express PD-1. Hence, the response of patients with cancer to PD-1 blockade therapy is a cumulative effect of anti-PD-1 antibodies acting on a myriad of cell types. Although, the contribution of CD8 T cells to PD-1 blockade therapy has been well-established, recent studies also suggest the involvement of non-canonical PD-1 signaling in blockade therapy. This review discusses the role of non-canonical PD-1 signaling in distinct cell types and explores how the available knowledge can improve PD-1 blockade immunotherapy, particularly in identifying novel biomarkers and combination treatment strategies.
Collapse
Affiliation(s)
- Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Ying Jiang
- Postgraduate Training Base in Rocket Army Special Medical Center of the PLA, Jinzhou Medical University, Jinzhou, P.R. China
| | - Xi Wang
- Otorhinolaryngology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Jin Shang
- Department of Health Service, Guard Bureau of the Joint Staff Department, Central Military Commission of PLA, Beijing, P.R. China
| | - Ning Wang
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Lei Yu
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Wei Zhao
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Zhihua Li
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Juan An
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Xiaochun Zhang
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Huoming Chen
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Zhaoxia Li
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| |
Collapse
|
35
|
Beck Enemark M, Monrad I, Madsen C, Lystlund Lauridsen K, Honoré B, Plesner TL, Hamilton-Dutoit SJ, d'Amore F, Ludvigsen M. PD-1 Expression in Pre-Treatment Follicular Lymphoma Predicts the Risk of Subsequent High-Grade Transformation. Onco Targets Ther 2021; 14:481-489. [PMID: 33500624 PMCID: PMC7822223 DOI: 10.2147/ott.s289337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Follicular lymphoma (FL) is an indolent, yet generally incurable neoplasia with a median survival exceeding 10 years. However, a subset of FL patients experiences histological transformation (HT) to a more aggressive lymphoma, in the majority of cases to diffuse large B-cell lymphoma (DLBCL). This affects both the clinical course and the prognostic outcome, resulting in a markedly reduced survival after transformation. Thus, early risk stratification and prediction of patients at risk of HT would be highly valuable in the clinical setting. Here, we investigated the potential of the immune inhibitory programmed death 1 (PD-1) receptor as a biomarker predictive of HT. Patients and Methods Immunohistochemical staining and quantification by digital image analysis of PD-1 was performed on diagnostic tumor-tissue samples from FL patients with and without subsequent transformation (n=34 and n=46, respectively), and on paired samples from the transformed lymphoma (n=34). Results At the time of initial FL diagnosis, samples from patients with subsequent HT had significantly higher tumor-tissue expression of PD-1 compared with diagnostic FL samples from patients without subsequent HT (p=0.010). At the time of transformation, PD-1 expression was significantly reduced (p<0.001). No difference was observed in intra-follicular PD-1 expression at FL diagnosis between samples from patients with or without HT; however, high intra-follicular levels of PD-1 were associated with significantly shorter transformation-free survival times (p<0.043). Conclusion Our data suggest that pre-treatment tumor-tissue PD-1 expression already predicts the risk of subsequent transformation to DLBCL, as early as the time of FL diagnosis.
Collapse
Affiliation(s)
- Marie Beck Enemark
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ida Monrad
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Charlotte Madsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Bent Honoré
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Francesco d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Maja Ludvigsen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
36
|
Romero-Olmedo AJ, Schulz AR, Huber M, Brehm CU, Chang HD, Chiarolla CM, Bopp T, Skevaki C, Berberich-Siebelt F, Radbruch A, Mei HE, Lohoff M. Deep phenotypical characterization of human CD3 + CD56 + T cells by mass cytometry. Eur J Immunol 2020; 51:672-681. [PMID: 33231295 DOI: 10.1002/eji.202048941] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/25/2020] [Accepted: 11/19/2020] [Indexed: 12/25/2022]
Abstract
CD56+ T cells are a group of pro-inflammatory CD3+ lymphocytes with characteristics of natural killer cells, being involved in antimicrobial immune defense. Here, we performed deep phenotypic profiling of CD3+ CD56+ cells in peripheral blood of normal human donors and individuals sensitized to birch-pollen or/and house dust mite by high-dimensional mass cytometry combined with manual and computational data analysis. A co-regulation between major conventional T-cell subsets and their respective CD3+ CD56+ cell counterparts appeared restricted to CD8+ , MAIT, and TCRγδ+ T-cell compartments. Interestingly, we find a co-regulation of several CD3+ CD56+ cell subsets in allergic but not in healthy individuals. Moreover, using FlowSOM, we distinguished a variety of CD56+ T-cell phenotypes demonstrating a hitherto underestimated heterogeneity among these cells. The novel CD3+ CD56+ subset description comprises phenotypes superimposed with naive, memory, type 1, 2, and 17 differentiation stages, in part represented by a phenotypical continuum. Frequencies of two out of 19 CD3+ CD56+ FlowSOM clusters were significantly diminished in allergic individuals, demonstrating less frequent presence of cells with cytolytic, presumably protective, capacity in these donors consistent with defective expansion or their recruitment to the affected tissue. Our results contribute to defining specific cell populations to be targeted during therapy for allergic conditions.
Collapse
Affiliation(s)
- Addi J Romero-Olmedo
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Axel R Schulz
- German Rheumatism Research Center Berlin (DRFZ), Leibniz Institute, Berlin, Germany
| | - Magdalena Huber
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| | - Corinna U Brehm
- Comprehensive Biobank Marburg - CBBMR, Member of the DZL, Philipps-University Marburg, Marburg, Germany.,Institute for Pathology, University Hospital Marburg, Philipps-University Marburg, Marburg, Germany
| | - Hyun-Dong Chang
- German Rheumatism Research Center Berlin (DRFZ), Leibniz Institute, Berlin, Germany
| | - Cristina M Chiarolla
- Institute of Pathology, Julius-Maximilian University of Wuerzburg, Wuerzburg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Chrysanthi Skevaki
- Institute of Laboratory Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | | | - Andreas Radbruch
- German Rheumatism Research Center Berlin (DRFZ), Leibniz Institute, Berlin, Germany
| | - Henrik E Mei
- German Rheumatism Research Center Berlin (DRFZ), Leibniz Institute, Berlin, Germany
| | - Michael Lohoff
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, Germany
| |
Collapse
|
37
|
Wang SR, Zhong N, Zhang XM, Zhao ZB, Balderas R, Li L, Lian ZX. OMIP 071: A 31-Parameter Flow Cytometry Panel for In-Depth Immunophenotyping of Human T-Cell Subsets Using Surface Markers. Cytometry A 2020; 99:273-277. [PMID: 33219622 DOI: 10.1002/cyto.a.24272] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022]
Abstract
Dissecting the functional diversity of T cells is critical in elucidating mechanisms and in developing therapies for various diseases. Here, we designed a 31-parameter (29-color) panel to enable the characterization of T-cell subsets and immunophenotyping of the human peripheral blood and lymph nodes using cell surface staining. In addition to adaptive T-cell markers, TCR Vα24-Jα18, TCR γδ, TCR Vɑ7.2, and CD161 were included to identify iNKT, γδ T, and MAIT cells, respectively, which are innate-like T cells. C-X-C chemokine receptors (CXCR3, CXCR4, CXCR5, CXCR6) and C-C motif chemokine receptors (CCR4, CCR6, CCR7) were included to enable the identification of Th cell subsets (Th1, Th2, Th17), Tfh cell subsets (Tfh1, Tfh2, Tfh17), and Th cells with specific homing capacities. Furthermore, in this panel, we also used markers for assessing cell differentiation (CD45RO, CD7), activation (CD57, CD95, HLA-DR) and the expression of some cosignaling molecules (PD-1, NKG2D, CD28). Particularly, CD69 and CD103 were included for the further analysis of tissue resident memory T (Trm) cells. This panel would enable the in-depth immunophenotyping of human T-cell subsets, and may be applied in the monitoring, prognosis, and mechanistic studies of various immune-related diseases.
Collapse
Affiliation(s)
- Song-Rong Wang
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.,Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.,Center of Excellence, Becton Dickinson Medical Devices (Shanghai) Co., Ltd and South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Na Zhong
- Center of Excellence, Becton Dickinson Medical Devices (Shanghai) Co., Ltd and South China University of Technology, Guangzhou, Guangdong, 510006, China.,Becton Dickinson Medical Devices (Shanghai) Co., Ltd, Guangzhou, Guangdong, 510180, China
| | - Xin-Mei Zhang
- Center of Excellence, Becton Dickinson Medical Devices (Shanghai) Co., Ltd and South China University of Technology, Guangzhou, Guangdong, 510006, China.,Becton Dickinson Medical Devices (Shanghai) Co., Ltd, Guangzhou, Guangdong, 510180, China
| | - Zhi-Bin Zhao
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.,Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.,Center of Excellence, Becton Dickinson Medical Devices (Shanghai) Co., Ltd and South China University of Technology, Guangzhou, Guangdong, 510006, China
| | | | - Liang Li
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.,Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.,Center of Excellence, Becton Dickinson Medical Devices (Shanghai) Co., Ltd and South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Zhe-Xiong Lian
- Department of General Surgery, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China.,Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.,Center of Excellence, Becton Dickinson Medical Devices (Shanghai) Co., Ltd and South China University of Technology, Guangzhou, Guangdong, 510006, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510005, China
| |
Collapse
|
38
|
Ansell SM. Fundamentals of immunology for understanding immunotherapy for lymphoma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:585-589. [PMID: 33275742 PMCID: PMC7727535 DOI: 10.1182/hematology.2020002537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
An effective antitumor immune response in patients with lymphoma would eradicate the malignant B cells and cure the patient of the disease. This, however, does not occur, and a suboptimal antitumor response results in persistence and subsequent progression of the patient's disease. The goals of immunotherapy are therefore to restore an effective antitumor immune response by promoting immune recognition, optimizing immune activation, and supporting persistence of the immune response resulting in subsequent immunological memory. Multiple mechanisms, however, are present within the tumor microenvironment that account for an inadequate immune response. These include loss of major histocompatibility complex expression on tumor cells and subsequent inadequate antigen presentation, increased expression of immunosuppressive ligands on malignant cells, populations of immune cells with suppressive function present in the tumor, and cytokines secreted by the malignant cell or other cells in the microenvironment that promote immune exhaustion or suppress the immune response. Successful immunotherapeutic strategies are specifically addressing these issues by promoting antigen presentation, improving recognition of the malignant cell, directly activating T cells and natural killer cells, and blocking immune checkpoint signaling that would suppress the immune response. Many of these approaches have proven highly successful in patients with various subtypes of lymphoma and are now being incorporated into standard clinical practice.
Collapse
|
39
|
Ansell SM. Fundamentals of immunology for understanding immunotherapy for lymphoma. Blood Adv 2020; 4:5863-5867. [PMID: 33232478 PMCID: PMC7686892 DOI: 10.1182/bloodadvances.2020002537] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/11/2020] [Indexed: 02/08/2023] Open
Abstract
An effective antitumor immune response in patients with lymphoma would eradicate the malignant B cells and cure the patient of the disease. This, however, does not occur, and a suboptimal antitumor response results in persistence and subsequent progression of the patient's disease. The goals of immunotherapy are therefore to restore an effective antitumor immune response by promoting immune recognition, optimizing immune activation, and supporting persistence of the immune response resulting in subsequent immunological memory. Multiple mechanisms, however, are present within the tumor microenvironment that account for an inadequate immune response. These include loss of major histocompatibility complex expression on tumor cells and subsequent inadequate antigen presentation, increased expression of immunosuppressive ligands on malignant cells, populations of immune cells with suppressive function present in the tumor, and cytokines secreted by the malignant cell or other cells in the microenvironment that promote immune exhaustion or suppress the immune response. Successful immunotherapeutic strategies are specifically addressing these issues by promoting antigen presentation, improving recognition of the malignant cell, directly activating T cells and natural killer cells, and blocking immune checkpoint signaling that would suppress the immune response. Many of these approaches have proven highly successful in patients with various subtypes of lymphoma and are now being incorporated into standard clinical practice.
Collapse
|
40
|
Gonder S, Fernandez Botana I, Wierz M, Pagano G, Gargiulo E, Cosma A, Moussay E, Paggetti J, Largeot A. Method for the Analysis of the Tumor Microenvironment by Mass Cytometry: Application to Chronic Lymphocytic Leukemia. Front Immunol 2020; 11:578176. [PMID: 33193376 PMCID: PMC7606286 DOI: 10.3389/fimmu.2020.578176] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/29/2020] [Indexed: 12/21/2022] Open
Abstract
In the past 20 years, the interest for the tumor microenvironment (TME) has exponentially increased. Indeed, it is now commonly admitted that the TME plays a crucial role in cancer development, maintenance, immune escape and resistance to therapy. This stands true for hematological malignancies as well. A considerable amount of newly developed therapies are directed against the cancer-supporting TME instead of targeting tumor cells themselves. However, the TME is often not clearly defined. In addition, the unique phenotype of each tumor and the variability among patients limit the success of such therapies. Recently, our group took advantage of the mass cytometry technology to unveil the specific TME in the context of chronic lymphocytic leukemia (CLL) in mice. We found the enrichment of LAG3 and PD1, two immune checkpoints. We tested an antibody-based immunotherapy, targeting these two molecules. This combination of antibodies was successful in the treatment of murine CLL. In this methods article, we provide a detailed protocol for the staining of CLL TME cells aiming at their characterization using mass cytometry. We include panel design and validation, sample preparation and acquisition, machine set-up, quality control, and analysis. Additionally, we discuss different advantages and pitfalls of this technique.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Flow Cytometry
- Immune Checkpoint Inhibitors/pharmacology
- Immunotherapy
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Mice
- Programmed Cell Death 1 Receptor/metabolism
- Tumor Microenvironment
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Susanne Gonder
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Iria Fernandez Botana
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marina Wierz
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Giulia Pagano
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Ernesto Gargiulo
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Antonio Cosma
- National Cytometry Platform, Quantitative Biology Unit, Transversal Activities, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Etienne Moussay
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Jerome Paggetti
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Anne Largeot
- Tumor Stroma Interactions, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| |
Collapse
|
41
|
Zhang T, Warden AR, Li Y, Ding X. Progress and applications of mass cytometry in sketching immune landscapes. Clin Transl Med 2020; 10:e206. [PMID: 33135337 PMCID: PMC7556381 DOI: 10.1002/ctm2.206] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Recently emerged mass cytometry (cytometry by time-of-flight [CyTOF]) technology permits the identification and quantification of inherently diverse cellular systems, and the simultaneous measurement of functional attributes at the single-cell resolution. By virtue of its multiplex ability with limited need for compensation, CyTOF has led a critical role in immunological research fields. Here, we present an overview of CyTOF, including the introduction of CyTOF principle and advantages that make it a standalone tool in deciphering immune mysteries. We then discuss the functional assays, introduce the bioinformatics to interpret the data yield via CyTOF, and depict the emerging clinical and research applications of CyTOF technology in sketching immune landscape in a wide variety of diseases.
Collapse
Affiliation(s)
- Ting Zhang
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Antony R. Warden
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yiyang Li
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Xianting Ding
- State Key laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
42
|
Abstract
Histologic transformation of follicular lymphoma remains the leading cause of follicular lymphoma-related mortality in the rituximab era. Both the diverse timing of transformation and heterogeneity in associated genomic events suggest that histologic transformation may itself comprise distinct disease entities. Successive indolent and transformation episodes occur by divergent clonal evolution from an inferred common progenitor cell, representing a potential therapeutic target. Existing biological knowledge largely pre-dates anti-CD20 therapy, and further prospective validation is essential. Inclusion of transformation cases in clinical trials incorporating biomarker discovery, and an integrated understanding of the genetic and microenvironmental factors underpinning transformation, may unearth renewed clinical opportunities.
Collapse
Affiliation(s)
- Emil A Kumar
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Jessica Okosun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jude Fitzgibbon
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. https://twitter.com/fitzgi02
| |
Collapse
|
43
|
Single-cell mass cytometry on peripheral blood identifies immune cell subsets associated with primary biliary cholangitis. Sci Rep 2020; 10:12584. [PMID: 32724082 PMCID: PMC7387528 DOI: 10.1038/s41598-020-69358-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/07/2020] [Indexed: 02/08/2023] Open
Abstract
The relationship between primary biliary cholangitis (PBC), a chronic cholestatic autoimmune liver disease, and the peripheral immune system remains to be fully understood. Herein, we performed the first mass cytometry (CyTOF)-based, immunophenotyping analysis of the peripheral immune system in PBC at single-cell resolution. CyTOF was performed on peripheral blood mononuclear cells (PBMCs) from PBC patients (n = 33) and age-/sex-matched healthy controls (n = 33) to obtain immune cell abundance and marker expression profiles. Hierarchical clustering methods were applied to identify immune cell types and subsets significantly associated with PBC. Subsets of gamma-delta T cells (CD3+TCRgd+), CD8+ T cells (CD3+CD8+CD161+PD1+), and memory B cells (CD3−CD19+CD20+CD24+CD27+) were found to have lower abundance in PBC than in control. In contrast, higher abundance of subsets of monocytes and naïve B cells were observed in PBC compared to control. Furthermore, several naïve B cell (CD3−CD19+CD20+CD24−CD27−) subsets were significantly higher in PBC patients with cirrhosis (indicative of late-stage disease) than in those without cirrhosis. Alternatively, subsets of memory B cells were lower in abundance in cirrhotic relative to non-cirrhotic PBC patients. Future immunophenotyping investigations could lead to better understanding of PBC pathogenesis and progression, and also to the discovery of novel biomarkers and treatment strategies.
Collapse
|
44
|
Yang ZZ, Kim HJ, Wu H, Jalali S, Tang X, Krull JE, Ding W, Novak AJ, Ansell SM. TIGIT Expression Is Associated with T-cell Suppression and Exhaustion and Predicts Clinical Outcome and Anti-PD-1 Response in Follicular Lymphoma. Clin Cancer Res 2020; 26:5217-5231. [PMID: 32631956 DOI: 10.1158/1078-0432.ccr-20-0558] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/01/2020] [Accepted: 06/30/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE T-cell immunoglobulin and ITIM domain (TIGIT), a member of the immune checkpoint family, is important in normal T-cell biology. However, the phenotypical profile and clinical relevance of TIGIT in follicular lymphoma is largely unknown. EXPERIMENTAL DESIGN Biopsy specimens from a cohort of 82 patients with follicular lymphoma were analyzed using mass cytometry to explore the phenotype and biological and clinical significance of TIGIT+ T cells. RESULTS TIGIT is highly expressed on intratumoral T cells and its expression alters T-cell phenotype in follicular lymphoma. TIGIT is abundantly expressed on Treg cells, resulting in an enhanced suppressive property. TIGIT expression on non-Treg/TFH T cells defines a population that exhibits an exhausted phenotype. Clinically, increased numbers of TIGIT+ T cells are associated with inferior patient outcomes and poor survival. We observe that anti-PD-1 therapy with pembrolizumab alters the phenotype of TIGIT+ T subsets and identifies a role for CD28 expression on TIGIT+ T cells in treatment response. CONCLUSIONS The current study provides a comprehensive analysis of the phenotypic profile of intratumoral TIGIT+ T subsets and their prognostic relevance in follicular lymphoma. Inhibition of TIGIT signaling may be an additional mechanism to prevent T-cell suppression and exhaustion in B-cell lymphoma.
Collapse
Affiliation(s)
- Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota.
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Hongyan Wu
- Department of Immunology, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jordan E Krull
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Wei Ding
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
45
|
Mintz MA, Cyster JG. T follicular helper cells in germinal center B cell selection and lymphomagenesis. Immunol Rev 2020; 296:48-61. [PMID: 32412663 PMCID: PMC7817257 DOI: 10.1111/imr.12860] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
Germinal centers (GCs) are confined anatomic regions where rapidly proliferating B cells undergo somatic mutation and selection and eventual differentiation into memory B cells or long-lived plasma cells. GCs are also the origin of malignancy, namely follicular lymphoma (FL), GC B cell-diffuse large B cell lymphoma (GCB-DLBCL), and Burkitt lymphoma (BL). GC B cell lymphomas maintain their GC transcriptional signatures and sustain many features of the GC microenvironment, including CD4+ T follicular helper (Tfh) cells. Tfh cells are essential for the formation and maintenance of GCs, providing critical helper signals such as CD40L. Large-scale sequencing efforts have led to new insights about the tightly regulated selection mechanisms that are commonly targeted during GC B cell lymphomagenesis. For instance, HVEM, a frequently mutated surface molecule in GC-derived lymphomas, engages the inhibitory receptor BTLA on Tfh cells and loss of HVEM leads to exaggerated T cell help. Here, we review current understanding of how Tfh cells contribute to the selection of GC B cells, with a particular emphasis on how Tfh cell signals may contribute to lymphomagenesis. The possibility of targeting Tfh cells for the treatment of GC-derived lymphomas is discussed.
Collapse
Affiliation(s)
- Michelle A Mintz
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| | - Jason G Cyster
- Department of Microbiology and Immunology, Howard Hughes Medical Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
46
|
Szumera-Ciećkiewicz A, Poleszczuk J, Kuczkiewicz-Siemion O, Paszkiewicz-Kozik E, Rymkiewicz G, Sokół K, Borysiuk A, Kotarska M, Kawecka M, Owczarek D, Pytlak B, Walewski J, Prochorec-Sobieszek M. PD1 distribution pattern, regardless of the cell origin, is an independent microenvironmental prognostic factor for progression-free survival in follicular lymphoma. Pathol Res Pract 2020; 216:153096. [PMID: 32853965 DOI: 10.1016/j.prp.2020.153096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 02/08/2023]
Abstract
Follicular lymphoma (FL) is a well-studied microenvironment-dependent hematological malignancy, but the crosstalk between various involved cell subtypes is still not fully understood. Recent promising results of immunotherapy in recurrent FL warrant the need for an in-depth analysis of the expression and role of immune system-related proteins in the FL microenvironment. Seventy-one patients with FL and available diagnostic paraffin blocks were enrolled in the retrospective analysis. Histopathological diagnoses were revised according to the World Health Organization recommendations. Patients were either observed (watch and wait/W&W group) or immediately treated with chemo(immuno)therapy regimens according to their clinical status. Immunohistochemical assessment of PD1, PDL1, CD4, CD8, CD163, CD68-KP1, CD68-PGM1 was performed. The scoring methods included both semi-quantitative estimation of positive cells and architectural pattern distribution. The differences between PD1 staining distribution and intensity were classified as intra/perifollicular vs. interfollicular/diffuse cells and presented bright vs. dim immunoreactivity, respectively. No statistically significant differences in the density distribution of the immunohistochemical stainings were found between W&W and chemo(immuno)therapy groups. Interfollicular/diffuse pattern of PD1 expression had significantly decreased progression-free survival when analyzing the whole cohort and patients on chemo(immuno)therapy (p = 0.014 and p = 0.07, respectively). The high dependence was not significant in the W&W group. PD1 positivity of cells did not correlate with CD4 or CD8 immunophenotype. Morphologically FL neoplastic cells were entirely PDL1 negative, but granular and membranous staining was detected in the FL microenvironment. In line with previous studies, PD1/PDL1 expression was predominantly localized in the FL microenvironment, indicating that FL cells might not be the direct target for anti-PDL1 therapy. However, we show that the localization of PD1 expression could be a viable progression-free survival biomarker for FL.
Collapse
Affiliation(s)
- Anna Szumera-Ciećkiewicz
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland.
| | - Jan Poleszczuk
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Poland; Department of Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Olga Kuczkiewicz-Siemion
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Ewa Paszkiewicz-Kozik
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Grzegorz Rymkiewicz
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Flow Cytometry Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Kamil Sokół
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Anita Borysiuk
- Flow Cytometry Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Martyna Kotarska
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Monika Kawecka
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Daria Owczarek
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Beata Pytlak
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Jan Walewski
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Monika Prochorec-Sobieszek
- Pathology Laboratory, Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| |
Collapse
|
47
|
Xie M, Jiang Q, Zhao S, Zhao J, Ye X, Qian W. Prognostic value of tissue-infiltrating immune cells in tumor microenvironment of follicular lymphoma: A meta-analysis. Int Immunopharmacol 2020; 85:106684. [PMID: 32540726 DOI: 10.1016/j.intimp.2020.106684] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The follicular lymphoma (FL) microenvironment is composed of follicular dendritic cells (FDCs), tumor-infiltrating CD4/CD8+ T cells (TILs), follicular regulatory T (Treg) cells, lymphoma-associated macrophages (LAMs), and immune checkpoint-related immune cells, all of which are relevant in the prognosis of FL, but their results remain controversial. Therefore, we performed this systematic review to explore the relationship between the FL microenvironment and prognosis. METHODS Relevant studies were identified from PubMed, EMBASE and the Cochrane Library. Twenty-three trials involving 3336 patients with FL were included for analysis. RESULTS This meta-analysis confirmed the unfavorable prognostic role of high CD21+/CD23+ FDC density in overall survival (OS) and progression-free survival (PFS). CD8+ or granzyme B+ TILs instead of CD4+ TILs are indicators for good OS. FoxP3+ Treg cells was not associated with prognosis, and even in subgroup analysis neither the number of cells nor the infiltration pattern had predictive value. A high degree of CD68+ macrophage infiltration was a negative prognostic factor for OS, but was associated with good prognosis in the rituximab-era subgroup. Although there was no correlation between PD1-positive immune cells and prognosis, subtypes with the follicular helper T (TFH) or exhausted T cell (TEX) phenotype tended to influence prognosis. The HR in the short time to transformation (TTT) analyses suggested that high CD68+ LAM numbers, diffuse pattern of FOXP3+ Treg cells and PD1+ cells, and high PD-L1 cell numbers are adverse factors leading to early transformation. CONCLUSIONS Multiple tissue-infiltratingimmune cells in microenvironment play critical and different roles in FL prognosis.
Collapse
Affiliation(s)
- Mixue Xie
- Department of Haematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Qi Jiang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Shuqi Zhao
- Department of Haematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Jing Zhao
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China
| | - Xiujin Ye
- Department of Haematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| | - Wenbin Qian
- Department of Haematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, China.
| |
Collapse
|
48
|
Kawashima A, Kanazawa T, Kidani Y, Yoshida T, Hirata M, Nishida K, Nojima S, Yamamoto Y, Kato T, Hatano K, Ujike T, Nagahara A, Fujita K, Morimoto-Okazawa A, Iwahori K, Uemura M, Imamura R, Ohkura N, Morii E, Sakaguchi S, Wada H, Nonomura N. Tumour grade significantly correlates with total dysfunction of tumour tissue-infiltrating lymphocytes in renal cell carcinoma. Sci Rep 2020; 10:6220. [PMID: 32277125 PMCID: PMC7148296 DOI: 10.1038/s41598-020-63060-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 03/21/2020] [Indexed: 02/08/2023] Open
Abstract
It is important to evaluate the clinical importance of both CD8 T cells and CD4 T cells expression simultaneously because they have crucial networks in tumour targeting immune responses. In 97 RCC patients, RNA sequencing and gene set enrichment analysis of both CD8 and CD4 T cells based on the expression levels of PD-1 and TIM-3 implied that the populations of PD-1+TIM-3+ CD8 T cells and PD-1lowTIM-3 + CD4 T cells were characterized as exhausted CD8 T cells and regulatory CD4 T cells, respectively. These populations of CD4 and CD8 T cells were significantly upregulated in the patients with RCC of higher WHO/ISUP grade (grades 3, 4) (P < 0.001). Moreover, the cytokine productivities of each population in both CD4 and CD8 T cells of the higher-grade patients were significantly lower than those of the lower-grade patients (P < 0.05). Multivariate analysis showed the prognosis of patients with metastatic RCC of higher WHO/ISUP grade treated by nivolumab to be significantly worse than that of patients with lower grade (P = 0.026). This study showed that tumour grade significantly correlated with dysfunction of both CD4+ and CD8+ TILs and the efficacy of nivolumab treatment.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/mortality
- Cytokines/metabolism
- Female
- Follow-Up Studies
- Hepatitis A Virus Cellular Receptor 2/metabolism
- Humans
- Kidney/pathology
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/immunology
- Kidney Neoplasms/mortality
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Middle Aged
- Neoplasm Grading
- Nivolumab/pharmacology
- Nivolumab/therapeutic use
- Prognosis
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/metabolism
- Progression-Free Survival
- RNA-Seq
- Retrospective Studies
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Treatment Outcome
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Atsunari Kawashima
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Takayuki Kanazawa
- Department of Clinical Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Yujiro Kidani
- Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Basic Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tetsuya Yoshida
- Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Basic Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Michinari Hirata
- Department of Clinical Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Kentaro Nishida
- Department of Clinical Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yoshiyuki Yamamoto
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Taigo Kato
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Koji Hatano
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takeshi Ujike
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Akira Nagahara
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kazutoshi Fujita
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Akiko Morimoto-Okazawa
- Department of Clinical Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kota Iwahori
- Department of Clinical Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Motohide Uemura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Urological Immuno-Oncology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Ryoichi Imamura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naganari Ohkura
- Department of Basic Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shimon Sakaguchi
- Department of Experimental Immunology, Immunology Frontier Research Centre, Osaka University, Suita, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumour Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
49
|
|
50
|
Roussel M, Lhomme F, Roe CE, Bartkowiak T, Gravelle P, Laurent C, Fest T, Irish JM. Mass cytometry defines distinct immune profile in germinal center B-cell lymphomas. Cancer Immunol Immunother 2020; 69:407-420. [PMID: 31919622 PMCID: PMC7764565 DOI: 10.1007/s00262-019-02464-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/26/2019] [Indexed: 02/08/2023]
Abstract
Tumor-associated macrophage and T-cell subsets are implicated in the pathogenesis of diffuse large B-cell lymphoma, follicular lymphoma, and classical Hodgkin lymphoma. Macrophages provide essential mechanisms of tumor immune evasion through checkpoint ligand expression and secretion of suppressive cytokines. However, normal and tumor-associated macrophage phenotypes are less well characterized than those of tumor-infiltrating T-cell subsets, and it would be especially valuable to know whether the polarization state of macrophages differs across lymphoma tumor microenvironments. Here, an established mass cytometry panel designed to characterize myeloid-derived suppressor cells and known macrophage maturation and polarization states was applied to characterize B-lymphoma tumors and non-malignant human tissue. High-dimensional single-cell analyses were performed using dimensionality reduction and clustering tools. Phenotypically distinct intra-tumor macrophage subsets were identified based on abnormal marker expression profiles that were associated with lymphoma tumor types. While it had been proposed that measurement of CD163 and CD68 might be sufficient to reveal macrophage subsets in tumors, results here indicated that S100A9, CCR2, CD36, Slan, and CD32 should also be measured to effectively characterize lymphoma-specific tumor macrophages. Additionally, the presence of phenotypically distinct, abnormal macrophage populations was closely linked to the phenotype of intra-tumor T-cell populations, including PD-1 expressing T cells. These results further support the close links between macrophage polarization and T-cell functional state, as well as the rationale for targeting tumor-associated macrophages in cancer immunotherapies.
Collapse
Affiliation(s)
- Mikael Roussel
- Laboratoire Hématologie, CHU Pontchaillou, Centre Hospitalier Universitaire de Rennes, Pôle Biologie, 2 rue Henri Le Guilloux, 35033, Rennes, France.
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France.
| | - Faustine Lhomme
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France
| | - Caroline E Roe
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Todd Bartkowiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Pauline Gravelle
- Service Anatomie et Cytologie Pathologiques and UMR1037, Toulouse, France
| | - Camille Laurent
- Service Anatomie et Cytologie Pathologiques and UMR1037, Toulouse, France
| | - Thierry Fest
- Laboratoire Hématologie, CHU Pontchaillou, Centre Hospitalier Universitaire de Rennes, Pôle Biologie, 2 rue Henri Le Guilloux, 35033, Rennes, France
- INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Equipe Labellisée Ligue Contre Le Cancer, Rennes, France
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, 740B Preston Building, 2220 Pierce Avenue, Nashville, TN, 37232-6840, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|