1
|
Menéndez V, Solórzano JL, García-Cosío M, Cereceda L, Díaz E, Estévez M, Roncador G, Vega Z, Montalbán C, Kulasinghe A, García JF. Mapping the Spatial Dynamics of the CD4+ T Cell Spectrum in Classical Hodgkin Lymphoma. Mod Pathol 2024; 37:100551. [PMID: 38936478 DOI: 10.1016/j.modpat.2024.100551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
As around 25% to 30% of classical Hodgkin lymphoma (cHL) patients with advanced stages do not respond to standard therapies, the tumor microenvironment of cHL is one avenue that may be explored with the aim of improving risk stratification. CD4+ T cells are thought to be one of the main cell types in the tumor microenvironment. However, few immune signatures have been studied, and many of these lack related spatial data. Thus, our aim is to spatially resolve the CD4+ T cell subtypes that influence cHL outcome, depicting new immune signatures or transcriptional patterns that are in crosstalk with the tumor cells. This study was conducted using the NanoString GeoMx digital spatial profiling technology, based on the selection of distinct functional areas of patients' tissues followed by gene-expression profiling. The goals were to assess the differences in CD4+ T cell populations between tumor-rich and immune-predominant areas defined by different CD30 and PD-L1 expression levels and seek correlations with clinical metadata. Our results depict a complex map of CD4+ T cells with different functions and differentiation states that are enriched at distinct locations, the flux of cytokines and chemokines that could be related to these, and the specific relationships with the clinical outcome.
Collapse
Affiliation(s)
- Victoria Menéndez
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain
| | - José L Solórzano
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain; Pathology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Mónica García-Cosío
- Department of Pathology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Laura Cereceda
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain; Pathology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Eva Díaz
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain
| | - Mónica Estévez
- Department of Hematology, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Giovanna Roncador
- Monoclonal Antibodies and Histopathology Units, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Zaira Vega
- Monoclonal Antibodies and Histopathology Units, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Carlos Montalbán
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain
| | - Arutha Kulasinghe
- Faculty of Medicine, Frazer Institute, The University of Queensland, Brisbane, Australia
| | - Juan F García
- Translational Research, MD Anderson Cancer Center Foundation, Madrid, Spain; Pathology Department, MD Anderson Cancer Center Madrid, Madrid, Spain.
| |
Collapse
|
2
|
Solórzano JL, Menéndez V, Parra E, Solis L, Salazar R, García-Cosío M, Climent F, Fernández S, Díaz E, Francisco-Cruz A, Khoury J, Jiang M, Tamegnon A, Montalbán C, Melero I, Wistuba I, De Andrea C, F. García J. Multiplex spatial analysis reveals increased CD137 expression and m-MDSC neighboring tumor cells in refractory classical Hodgkin Lymphoma. Oncoimmunology 2024; 13:2388304. [PMID: 39135889 PMCID: PMC11318683 DOI: 10.1080/2162402x.2024.2388304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
The Hodgkin and Reed - Sternberg (HRS) cells in classical Hodgkin Lymphoma (cHL) actively modify the immune tumor microenvironment (TME) attracting immunosuppressive cells and expressing inhibitory molecules. A high frequency of myeloid cells in the TME is correlated with an unfavorable prognosis, but more specific and rare cell populations lack precise markers. Myeloid-derived suppressor cells (MDSCs) have been identified in the peripheral blood of cHL patients, where they appear to be correlated with disease aggressiveness. TNFRSF9 (CD137) is a T cell co-stimulator expressed by monocytic and dendritic cells. Its expression has also been described in HRS cells, where it is thought to play a role in reducing antitumor responses. Here, we perform qualitative and quantitative analyses of lymphocytic and MDSC subtypes and determine the CD137 cell distribution in cHL primary tumors using multiplex immunofluorescence and automated multispectral imaging. The results were correlated with patients' clinical features. Cells were stained with specific panels of immune checkpoint markers (PD-1, PD-L1, CD137), tumor-infiltrating T lymphocytes (CD3, PD-1), and monocytic cells/MDSCs (CD68, CD14, CD33, Arg-1, CD11b). This approach allowed us to identify distinct phenotypes and to analyze spatial interactions between immune subpopulations and tumor cells. The results confirm CD137 expression by T, monocytic and HRS cells. In addition, the expression of CD137, T exhausted cells, and monocytic MDSCs (m-MDSCs) in the vicinity of malignant HRS cells were associated with a worse prognosis. Our findings reveal new elements of the TME that mediate immune escape, and confirm CD137 as a candidate target for immunotherapy in cHL.
Collapse
Affiliation(s)
- José L. Solórzano
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Victoria Menéndez
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Edwin Parra
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Luisa Solis
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | | | | | - Fina Climent
- Pathology Department, Hospital Universitari de Bellvitge, IDIBELL, L’Hospitalet De Llobregat, Barcelona, Spain
| | - Sara Fernández
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
| | - Eva Díaz
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | | | - Joseph Khoury
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mei Jiang
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Auriole Tamegnon
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Carlos Montalbán
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| | - Ignacio Melero
- University of Navarra and Instituto de Investigación Sanitaria de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, Houston, TX, USA
| | - Carlos De Andrea
- University of Navarra and Instituto de Investigación Sanitaria de Navarra, Pamplona, Navarra, Spain
| | - Juan F. García
- Pathology and Molecular Department, MD Anderson Cancer Center, Madrid, Spain
- Translational Research Department, MD Anderson Foundation, Madrid, Madrid, Spain
| |
Collapse
|
3
|
Yang P, Zang G, Yan Y, Zhong W, Li B, Xu Y, Shao C, Wang Z, Pu J, Yuan W. CD137-CD137L Aggravates Calcification of Vascular Smooth Muscle Cell and Vasculature of ApoE -/- Mice Via Rab7-Mediated Autophagy. J Cardiovasc Transl Res 2022; 15:1297-1314. [PMID: 35763154 DOI: 10.1007/s12265-022-10272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Vascular calcification is an independent risk factor for acute cardiovascular events and a predictor of adverse prognosis; the abnormal fusion and degradation of autophagosomes and lysosomes are closely related to the calcification of VSMC and aortic AS plaque in ApoE-/- mice. Rab7 is a member of the Ras protein family and acts as a molecular switch in the fusion between autophagosomes and lysosomes. In this study, we found that the activation of the CD137-CD137L signal promoted calcification by inhibiting the expression and activity of Rab7, which regulates the degradation of autophagic cargo in vascular smooth muscle cells (VSMCs) and aortic atherosclerosis (AS) plaques in ApoE-/- mice. Knockdown of Rab7 impaired its tethering with the downstream molecule FYVE and coiled-coil containing 1 (FYCO1), which transports autophagosomes to lysosomes through microtubule motor kinesins and fuses with lysosomes to degrade the autophagic content. Overexpression of Rab7-alleviated calcification caused by the activation of the CD137 signaling pathway. In addition, FYCO1 knockdown promoted calcification even though the expression and activity of Rab7 were normal. Our results suggest that Rab7 is the target of CD137 signaling; Rab7 cannot interact with its downstream molecule FYCO1 when its activity and expression were inhibited by the activation of CD137 signaling pathway, thus inhibiting the autophagic degradation and promoting calcification.
Collapse
Affiliation(s)
- Ping Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Guangyao Zang
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Yang Yan
- Department of Cardiology Ren Ji Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhong
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Bo Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Yao Xu
- School of Medicine, Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China
| | - Jun Pu
- Department of Cardiology Ren Ji Hospital Affiliated to Shanghai, Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Jiangsu Province, 212001, Zhenjiang, China.
| |
Collapse
|
4
|
The Hodgkin Lymphoma Immune Microenvironment: Turning Bad News into Good. Cancers (Basel) 2022; 14:cancers14051360. [PMID: 35267668 PMCID: PMC8909875 DOI: 10.3390/cancers14051360] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/09/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
The classic Hodgkin lymphoma (cHL) tumor microenvironment (TME) is by far the most abundant component of tumors and is responsible for most of their biological and clinical characteristics. Recent advances in our knowledge of these networks in cellular interactions allow us to understand that the neoplastic Hodgkin and Reed Sternberg (HRS) cells, although they are in the minority, are the main architects of this dysregulated immune milieu. Here, we review the major changes that have happened in recent years: from TME as a helpless bystander, reflecting an ineffective immune response, to a dynamic tumor-promoting and immunosuppressive element. The HRS cells promote survival through interconnected intrinsic and extrinsic alterations, boosting pro-tumoral signaling pathways through genetic aberrations and autocrine growth signals, in parallel with abnormal cytokine secretion for the recruitment and selection of the best cell partners for this immunosuppressive TME. In turn, cHL is already proving to be the perfect model with which to address an immune checkpoint blockade. Preliminary data demonstrate the utility of druggable key signaling pathways in this ensemble, such as JAK-STAT, NF-κB, and others. In addition, myriad biomarkers predicting a response await validation by new in situ multiplex analytical methods, single-cell gene expression, and other techniques. Together, these components will define the functional phenotypes with which we will elucidate the molecular pathogenesis of the disease and improve the survival of patients who are refractory to conventional therapies.
Collapse
|
5
|
Nanamori H, Sawada Y. Epigenetic Modification of PD-1/PD-L1-Mediated Cancer Immunotherapy against Melanoma. Int J Mol Sci 2022; 23:ijms23031119. [PMID: 35163049 PMCID: PMC8835029 DOI: 10.3390/ijms23031119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 02/06/2023] Open
Abstract
Malignant melanoma is one of the representative skin cancers with unfavorable clinical behavior. Immunotherapy is currently used for the treatment, and it dramatically improves clinical outcomes in patients with advanced malignant melanoma. On the other hand, not all these patients can obtain therapeutic efficacy. To overcome this limitation of current immunotherapy, epigenetic modification is a highlighted issue for clinicians. Epigenetic modification is involved in various physiological and pathological conditions in the skin. Recent studies identified that skin cancer, especially malignant melanoma, has advantages in tumor development, indicating that epigenetic manipulation for regulation of gene expression in the tumor can be expected to result in additional therapeutic efficacy during immunotherapy. In this review, we focus on the detailed molecular mechanism of epigenetic modification in immunotherapy, especially anti-PD-1/PD-L1 antibody treatment for malignant melanoma.
Collapse
|
6
|
Kassardjian A, Moatamed NA. Expression of immune checkpoint regulators, cytotoxic T lymphocyte antigen 4 (CTLA-4), and CD137 in cervical carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:1038-1047. [PMID: 34760040 PMCID: PMC8569305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/07/2021] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors have a significant role in oncology. One of these immune checkpoints is cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Inhibition of the CTLA-4 pathway has already led to the FDA approval of Ipilimumab (anti-CTLA-4), a targeted therapy for melanoma and other malignancies. CD137 is an inducible, costimulatory receptor of the tissue-necrosis-factor-receptor superfamily expressed on the activated immune cells. Clinical trials have also been set for anti-CD137 in several malignancies. We assessed CTLA-4 and CD137 expression on a tissue microarray (TMA) comprising of 99 core tissues which included normal, non-neoplastic, and neoplastic cervical lesions. When detected as strong granular cytoplasmic reaction in the epithelial cells, CTLA-4 expression was scored as positive. For CD137, the results were recorded based on the presence or absence of staining reaction on the cell membranes of the lymphoplasmacytic infiltrates. Overall, CTLA-4 was positive in 30% (30/100) of the cervical malignancies. Sub-categorically, 20% of invasive endocervical adenocarcinomas, 63% of adenosquamous carcinomas, and 31% of squamous cell carcinomas were positive for CTLA-4 with a tendency toward lower grade squamous cell carcinomas (SCCs). CD137 was positive in 100% lymphoplasmacytic infiltrates of endocervical adenocarcinomas, 90.5% of SCCs, and 87.5% of adenosquamous carcinomas. This study has found a significant expression of CTLA-4 in cervical cancer cells and CD137 positivity of lymphoplasmacytic infiltrates with potential for future targeted immunotherapy.
Collapse
Affiliation(s)
- Ari Kassardjian
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA Los Angeles 90095-1732, California, USA
| | - Neda A Moatamed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA Los Angeles 90095-1732, California, USA
| |
Collapse
|
7
|
Silva O, Charu V, Ewalt MD, Metcalf RA, Zhao S, Castellanos EM, Orellana E, Natkunam Y, Luna-Fineman S. Classic Hodgkin lymphoma in Guatemalan children of age less than six years: analysis of immune regulatory pathways and the tumor microenvironment. Leuk Lymphoma 2021; 62:1609-1618. [PMID: 33627023 DOI: 10.1080/10428194.2021.1885666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Classic Hodgkin lymphoma (cHL) in young children (ages 0-6) is rare in high income countries (HICs) but is more prevalent in low- and middle-income countries (LMICs) like Guatemala. Given that the majority of cHL studies have evaluated adolescent/adults, and the immune system changes with age, we sought to characterize Epstein-Barr virus (EBV) expression, immune regulatory pathway markers and the tumor microenvironment in 42 children ages 0-6 with cHL from Guatemala. We found a very high frequency of EBV expression (97.5%). Hodgkin cells showed increased expression of PD1 ligands and CD137, indicative of shared immune regulatory mechanisms with adult cHL. Pediatric cHL also showed an increase in CD8+ tumor infiltrating lymphocytes and tumor associated macrophages within the tumor microenvironment. Despite 25 having high risk disease, only 4 patients died from progressive disease, relapse or infection.
Collapse
Affiliation(s)
- Oscar Silva
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Vivek Charu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark D Ewalt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryan A Metcalf
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Shuchun Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Elizabeth Orellana
- Unidad Nacional de Oncologia Pediátrica, Guatemala City, Guatemala.,School of Medicine, Francisco Marroquin University, Guatemala City, Guatemala
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sandra Luna-Fineman
- Pediatric Hematology/Oncology/SCT, Center for Global Health, School of Medicine, University of Colorado Anschutz, Aurora, CO, USA
| |
Collapse
|
8
|
Lee KY, Wong HY, Zeng Q, Le Lin J, Cheng MS, Kuick CH, Chang KTE, Loh AHP, Schwarz H. Ectopic CD137 expression by rhabdomyosarcoma provides selection advantages but allows immunotherapeutic targeting. Oncoimmunology 2021; 10:1877459. [PMID: 33643694 PMCID: PMC7872024 DOI: 10.1080/2162402x.2021.1877459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 11/29/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is a heterogeneous soft tissue neoplasm most frequently found in children and adolescents. As the prognosis for recurrent and metastatic RMS remains poor, immunotherapies are hoped to improve quality of life and survival. CD137 is a member of tumor necrosis factor receptor family and a T cell costimulatory molecule which induces potent cellular immune responses that are able to eliminate malignant cells. Therefore, it was puzzling to find expression of CD137 on an RMS tissue microarray by multiplex staining. CD137 is not only expressed by infiltrating T cells but also by malignant RMS cells. Functional in vitro experiments demonstrate that CD137 on RMS cells is being transferred to adjacent antigen-presenting cells by trogocytosis, where it downregulates CD137 ligand, and thereby reduces T cell costimulation which results in reduced killing of RMS cells. The transfer of CD137 and the subsequent downregulation of CD137 ligand is a physiological negative feedback mechanism that is likely usurped by RMS, and may facilitate its escape from immune surveillance. In addition, CD137 signals into RMS cells and induces IL-6 and IL-8 secretion, which are linked to RMS metastasis and poor prognosis. However, the ectopic expression of CD137 on RMS cells is an Achilles' heel that may be utilized for immunotherapy. Natural killer cells expressing an anti-CD137 chimeric antigen receptor specifically kill CD137-expressing RMS cells. Our study implicates ectopic CD137 expression as a pathogenesis mechanism in RMS, and it demonstrates that CD137 may be a novel target for immunotherapy of RMS.
Collapse
Affiliation(s)
- Kang Yi Lee
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Hiu Yi Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Qun Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Jia Le Lin
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Man Si Cheng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | | | | | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
9
|
Poppema S. Lymphocyte predominant Hodgkin lymphoma, antigen-driven after all? J Pathol 2020; 253:1-10. [PMID: 33044742 DOI: 10.1002/path.5567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/22/2020] [Accepted: 10/06/2020] [Indexed: 01/12/2023]
Abstract
Nodular lymphocyte predominant Hodgkin lymphoma (NLPHL) was suggested as an entity separate from other types of Hodgkin lymphoma 40 years ago and recognized in the WHO classification in 2001. Based on its relatively benign course with late distant relapses, relation with lymph node hyperplasia with progressively transformed germinal centers, presence of clonal immunoglobulin gene rearrangements with somatic hypermutations and ongoing mutations, and relation with a number of inherited defects affecting the immune system, it has been suspected that NLPHL might be antigen-driven. Recent evidence has shown that cases of IgD-positive NLPHL are associated with infection by Moraxella catarrhalis, a common bacterium in the upper respiratory tract and in lymph nodes. This review summarizes the evidence for NLPHL as a B-cell lymphoma involving follicular T-lymphocytes normally found in germinal centers, its molecular features and relation to inherited immune defects, and its relation and differential diagnosis from similar entities. Finally, it discusses the evidence that in many cases a watch and wait policy might be a viable initial management strategy. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sibrandes Poppema
- School of Medical and Health Sciences, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
10
|
Shakerin P, Sedighi Moghadam B, Baghaei K, Safaei Naraghi Z, Kamyab Hesari K, Asadzadeh Aghdaei H, Shokouhi Shoormasti R, Fazeli MS, Nourizadeh M. Increasing the expression of programmed death ligand 2 (PD-L2) but not 4-1BB ligand in colorectal cancer cells. Mol Biol Rep 2020; 47:5689-5697. [PMID: 32661872 DOI: 10.1007/s11033-020-05289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/28/2020] [Indexed: 11/28/2022]
Abstract
Immune checkpoint (ICP) molecules modulate the immune response by either inducing or preventing T cell activation. Over-expression of some ICPs on malignant cells has been shown to regulate anti-tumor immune responses. We aimed to investigate the expression levels of two immune checkpoint molecules which have not been studied extensively in patients with colorectal cancer (CRC). Programmed Death Ligand 2 (co-inhibitory) and 4-1BB ligand (co-stimulatory) were assessed in tumor tissues of CRC patients compared to the adjacent normal tissues. Following tissue excision during surgical operation from 21 CRC patients, RNA extraction, cDNA synthesis and semi-quantitative real-time PCR were done for measuring the expressions of PD-L2 and 4-1BBL genes. In protein level, indirect immunohistochemistery (IHC) was performed on tissue sections. We revealed that PD-L2 was expressed in about 81% CRCs and insignificantly correlated with the tumor differentiation grade. Although a 3.25-fold change in the gene expression of PD-L2 was found in tumor tissues compared to the adjacent normal tissues (P = 0.005), but decreased level of 4-1BBL in counterpart tissues was not significant. Our results were confirmed by IHC for PDL-2 (P = 0.02) and 4-1BBL, however it was not statistically significant for the latter one. Although not significant, we could find an association between the elevated expression of PD-L2 and the tumor differentiation grade. Increased expression of negative regulator of the anti-tumor immune responses like PD-L2, as a prominent way of tumor escape, can be considered for cancer immunotherapy approaches in CRC patients using blocking monoclonal antibodies.
Collapse
Affiliation(s)
- Parastoo Shakerin
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Bijan Sedighi Moghadam
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Kaveh Baghaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Safaei Naraghi
- Department of Dermatopathology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Kambiz Kamyab Hesari
- Department of Dermatopathology, Razi Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Raheleh Shokouhi Shoormasti
- Immunology, Asthma, & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| | - Mohammad Sadegh Fazeli
- Department of Surgery, School of Medicine. Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Nourizadeh
- Immunology, Asthma, & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, 1419733151, Iran.
| |
Collapse
|
11
|
Zeng Q, Schwarz H. The role of trogocytosis in immune surveillance of Hodgkin lymphoma. Oncoimmunology 2020; 9:1781334. [PMID: 32934884 PMCID: PMC7466850 DOI: 10.1080/2162402x.2020.1781334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/28/2022] Open
Abstract
Hodgkin lymphoma (HL) is a unique type of hematopoietic cancer that has few tumor cells but a massive infiltration of immune cells. Findings on how the cancerous Hodgkin and Reed-Sternberg (HRS) cells survive and evade immune surveillance have facilitated the development of novel immunotherapies for HL. Trogocytosis is a fast process of intercellular transfer of membrane patches, which can significantly affect immune responses. In this review, we summarize the current knowledge of how trogocytosis contributes to the suppression of immune responses in HL. We focus on the ectopic expression of CD137 on HRS cells, the cause of its expression, and its implication on developing novel therapies for HL. Further, we review data demonstrating that similar mechanisms apply to CD30, PD-L1 and CTLA-4.
Collapse
Affiliation(s)
- Qun Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
12
|
Luu K, Shao Z, Schwarz H. The relevance of soluble CD137 in the regulation of immune responses and for immunotherapeutic intervention. J Leukoc Biol 2020; 107:731-738. [PMID: 32052477 DOI: 10.1002/jlb.2mr1119-224r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/10/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
CD137 is a potent costimulatory receptor. Several agonistic anti-CD137 antibodies are currently in clinical trials for tumor immunotherapy. Soluble forms of CD137 (sCD137) are generated by differential splicing and antagonize the activities of membrane-bound CD137 (mCD137) and of therapeutic CD137 agonists. sCD137 is found in sera of patients suffering from autoimmune diseases where it is a natural regulator of immune responses, and which has therapeutic potential for immune-mediated diseases. This review summarizes the current knowledge on sCD137, highlights its potential role in immunotherapy against cancer and in autoimmune diseases, and presents important issues to be addressed by future research.
Collapse
Affiliation(s)
- Khang Luu
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
| | - Zhe Shao
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
13
|
Luu K, Nickles E, Schwarz H. Destroy, what destroys you. Oncoimmunology 2019; 9:1685301. [PMID: 32002301 PMCID: PMC6959443 DOI: 10.1080/2162402x.2019.1685301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 11/20/2022] Open
Abstract
New evidence indicates the importance of CD137 for controlling Epstein-Barr virus (EBV) infections. (1) Mutations in CD137 predispose to EBV-associated diseases. (2) EBV induces ectopic CD137 expression, thereby activating a negative feed-back regulation and reducing T cell costimulation. These findings suggest CD137 agonists as new treatments for EBV-associated diseases.
Collapse
Affiliation(s)
- Khang Luu
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
| | - Emily Nickles
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
14
|
Rajendran S, Li Y, Ngoh E, Wong HY, Cheng MS, Wang CI, Schwarz H. Development of a Bispecific Antibody Targeting CD30 and CD137 on Hodgkin and Reed-Sternberg Cells. Front Oncol 2019; 9:945. [PMID: 31616638 PMCID: PMC6768943 DOI: 10.3389/fonc.2019.00945] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 09/09/2019] [Indexed: 01/12/2023] Open
Abstract
Hodgkin Lymphoma (HL) is a malignancy that frequently affects young adults. Although, there are effective treatments not every patient responds, necessitating the development of novel therapeutic approaches, especially for relapsed and refractory cases. The two TNF receptor family members CD30 and CD137 are expressed on Hodgkin and Reed Sternberg (HRS) cells, the malignant cells in HL. We found that this co-expression is specific for HRS cells. Based on this discovery we developed a bispecific antibody that binds preferentially to the CD30, CD137-double positive HRS cells. The CD30, CD137 bispecific antibody gets internalized into HRS cells opening up the possibility to use it as a carrier for a toxin. This antibody also induces antibody-dependent, cell-mediated cytotoxicity in CD30, CD137-double positive HRS cells. The enhances specificity of the CD30, CD137 bispecific antibody to HRS cells makes it a promising candidate for development as a novel HL treatment.
Collapse
Affiliation(s)
- Sakthi Rajendran
- Department of Physiology, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Yating Li
- Department of Physiology, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Evelyn Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hiu Yi Wong
- Department of Physiology, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Man Si Cheng
- Department of Physiology, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, National University of Singapore, Singapore, Singapore
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Aravinth SP, Rajendran S, Li Y, Wu M, Yi Wong AH, Schwarz H. Epstein-Barr virus-encoded LMP1 induces ectopic CD137 expression on Hodgkin and Reed-Sternberg cells via the PI3K-AKT-mTOR pathway. Leuk Lymphoma 2019; 60:2697-2704. [PMID: 31058559 DOI: 10.1080/10428194.2019.1607330] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CD137 is a potent co-stimulatory molecule on activated T cells, and its ligand (CD137L) is expressed on antigen presenting cells (APC). Ectopic expression of CD137 has been identified on Hodgkin Reed-Sternberg (HRS) cells, the malignant cells in Hodgkin Lymphoma (HL), and CD137 on HRS cells was found to support growth of HRS cells and escape from immune surveillance. HRS cells are mostly derived from B cells, which poses the question of how B cells acquire ectopic CD137 expression during the transformation process. HL is associated with Epstein-Barr virus (EBV) infection. We show that the EBV latent membrane protein 1 (LMP1) induces expression of CD137 in HRS cell lines. In a HL tissue microarray, 96% of the CD137-positive HL cases stained positive for LMP1. LMP1 utilizes the PI3K-AKT-mTOR pathway for inducing CD137 expression. These findings support the role of EBV in HL pathogenesis.
Collapse
Affiliation(s)
- Sneha Priya Aravinth
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sakthi Rajendran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Yating Li
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Meihui Wu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anna Hiu Yi Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
16
|
Wu M, Wong HY, Lin JL, Moliner A, Schwarz H. Induction of CD137 expression by viral genes reduces T cell costimulation. J Cell Physiol 2019; 234:21076-21088. [DOI: 10.1002/jcp.28710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/26/2019] [Accepted: 04/10/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Meihui Wu
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Hiu Yi Wong
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Jia Le Lin
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Annalena Moliner
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Herbert Schwarz
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| |
Collapse
|
17
|
Oshiro H, Tome Y, Kiyuna T, Miyake K, Kawaguchi K, Higuchi T, Miyake M, Zang Z, Razmjooei S, Barangi M, Wangsiricharoen S, Nelson SD, Li Y, Bouvet M, Singh SR, Kanaya F, Hoffman RM. Temozolomide targets and arrests a doxorubicin-resistant follicular dendritic-cell sarcoma patient-derived orthotopic xenograft mouse model. Tissue Cell 2019; 58:17-23. [PMID: 31133242 DOI: 10.1016/j.tice.2019.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/01/2019] [Indexed: 11/24/2022]
Abstract
Follicular dendritic cell sarcoma (FDCS) is a very rare and highly recalcitrant disease. A patient's doxorubicin-resistant FDCS was previously established orthotopically on the right high thigh into the biceps femoris of mice to establish a patient-derived orthotopic xenograft (PDOX) model. The aim of the present manuscript was to identify an effective drug for this recalcitrant tumor. Here, we evaluated the efficacy of temozolomide (TMZ), trabectedin (TRAB) and pazopanib (PAZ) on the FDCS PDOX model. PDOX mouse models were randomized into five groups of eight to nine mice, respectively. Group 1, untreated control with PBS, i.p.; Group 2, treated with doxorubicin (DOX), 2.4 mg/kg, i.p., weekly for 3 weeks; Group 3, treated with PAZ, 50 mg/kg, oral gavage, daily for 3 weeks; Group 4, treated with TMZ, 25 mg/kg, oral gavage, daily for 3 weeks; Group 5, treated with TRAB, 0.15 mg/kg, i.v., weekly for 3 weeks. Body weight and tumor volume were assessed 2 times per week. TMZ arrested the FDCS PDOX model compared to the control group (p < 0.05). PAZ and TRAB did not have significant efficacy compared to the control group (p = 0.99, p = 0.69 respectively). The PDOX tumor was resistant to DOX (p= 0.99). as was the patient. The present study demonstrates that TMZ is effective for a PDOX model of FDCS established from a patient who failed DOX treatment, further demonstrating the power of PDOX to identify effective therapy including for tumors that failed first line therapy.
Collapse
Affiliation(s)
- Hiromichi Oshiro
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yasunori Tome
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.
| | - Tasuku Kiyuna
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Kentaro Miyake
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Kei Kawaguchi
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Takashi Higuchi
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Masuyo Miyake
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Zhiying Zang
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Sahar Razmjooei
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Maryam Barangi
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Sintawat Wangsiricharoen
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Scott D Nelson
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Yunfeng Li
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, USA
| | - Shree Ram Singh
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| | - Fuminori Kanaya
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Robert M Hoffman
- AntiCancer Inc., San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
18
|
Carbone PN, Zhang QY. Validation of CD137 Immunohistochemical Stain on Paraffin-Embedded Tissue as a Marker to Facilitate Distinction Between Classic Hodgkin Lymphoma, Nodular Lymphocyte-Predominant Hodgkin Lymphoma, T-Cell/Histiocyte-Rich Large B-Cell Lymphoma, and Anaplastic Large Cell Lymphoma. Arch Pathol Lab Med 2019; 143:280-281. [PMID: 30816829 DOI: 10.5858/arpa.2018-0345-le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Peter N Carbone
- Department of Pathology, Naval Medical Center Portsmouth, Portsmouth, Virginia
| | - Qian-Yun Zhang
- Department of Pathology, The University of New Mexico School of Medicine/TriCore Reference Labs, Albuquerque
| |
Collapse
|
19
|
Gide TN, Quek C, Menzies AM, Tasker AT, Shang P, Holst J, Madore J, Lim SY, Velickovic R, Wongchenko M, Yan Y, Lo S, Carlino MS, Guminski A, Saw RPM, Pang A, McGuire HM, Palendira U, Thompson JF, Rizos H, Silva IPD, Batten M, Scolyer RA, Long GV, Wilmott JS. Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell 2019; 35:238-255.e6. [PMID: 30753825 DOI: 10.1016/j.ccell.2019.01.003] [Citation(s) in RCA: 633] [Impact Index Per Article: 105.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 11/07/2018] [Accepted: 01/07/2019] [Indexed: 11/21/2022]
Abstract
Cancer immunotherapies provide survival benefits in responding patients, but many patients fail to respond. Identifying the biology of treatment response and resistance are a priority to optimize drug selection and improve patient outcomes. We performed transcriptomic and immune profiling on 158 tumor biopsies from melanoma patients treated with anti-PD-1 monotherapy (n = 63) or combined anti-PD-1 and anti-CTLA-4 (n = 57). These data identified activated T cell signatures and T cell populations in responders to both treatments. Further mass cytometry analysis identified an EOMES+CD69+CD45RO+ effector memory T cell phenotype that was significantly more abundant in responders to combined immunotherapy compared with non-responders (n = 18). The gene expression profile of this population was associated with longer progression-free survival in patients treated with single agent and greater tumor shrinkage in both treatments.
Collapse
MESH Headings
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Drug Resistance, Neoplasm
- Female
- Humans
- Immunologic Memory/drug effects
- Ipilimumab/administration & dosage
- Lectins, C-Type/immunology
- Leukocyte Common Antigens/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Male
- Melanoma/drug therapy
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/pathology
- Middle Aged
- Nivolumab/administration & dosage
- Phenotype
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Retrospective Studies
- Signal Transduction/drug effects
- Skin Neoplasms/drug therapy
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Treatment Outcome
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Tuba N Gide
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; Department of Medical Oncology, Mater Hospital, North Sydney, NSW 2060, Australia
| | - Annie T Tasker
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia
| | - Ping Shang
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia
| | - Jeff Holst
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Centenary Institute, The University of Sydney, Sydney, NSW 2050, Australia
| | - Jason Madore
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia
| | - Su Yin Lim
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Rebecca Velickovic
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia
| | - Matthew Wongchenko
- Oncology Biomarker Development, Genentech Inc, South San Francisco, CA 94080, USA
| | - Yibing Yan
- Oncology Biomarker Development, Genentech Inc, South San Francisco, CA 94080, USA
| | - Serigne Lo
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, NSW 2145, Australia
| | - Alexander Guminski
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; Department of Medical Oncology, Mater Hospital, North Sydney, NSW 2060, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Angel Pang
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Centenary Institute, The University of Sydney, Sydney, NSW 2050, Australia
| | - Helen M McGuire
- Ramaciotti Facility for Human Systems Biology, Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia; Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Umaimainthan Palendira
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Centenary Institute, The University of Sydney, Sydney, NSW 2050, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Helen Rizos
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Ines Pires da Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Marcel Batten
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW 2065, Australia; Department of Medical Oncology, Mater Hospital, North Sydney, NSW 2060, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
20
|
Antigenic Targets for the Immunotherapy of Acute Myeloid Leukaemia. J Clin Med 2019; 8:jcm8020134. [PMID: 30678059 PMCID: PMC6406328 DOI: 10.3390/jcm8020134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/10/2019] [Accepted: 01/20/2019] [Indexed: 12/18/2022] Open
Abstract
One of the most promising approaches to preventing relapse is the stimulation of the body’s own immune system to kill residual cancer cells after conventional therapy has destroyed the bulk of the tumour. In acute myeloid leukaemia (AML), the high frequency with which patients achieve first remission, and the diffuse nature of the disease throughout the periphery, makes immunotherapy particularly appealing following induction and consolidation therapy, using chemotherapy, and where possible stem cell transplantation. Immunotherapy could be used to remove residual disease, including leukaemic stem cells from the farthest recesses of the body, reducing, if not eliminating, the prospect of relapse. The identification of novel antigens that exist at disease presentation and can act as targets for immunotherapy have also proved useful in helping us to gain a better understand of the biology that belies AML. It appears that there is an additional function of leukaemia associated antigens as biomarkers of disease state and survival. Here, we discuss these findings.
Collapse
|
21
|
Aberrant CD137 ligand expression induced by GATA6 overexpression promotes tumor progression in cutaneous T-cell lymphoma. Blood 2018; 132:1922-1935. [PMID: 30194255 DOI: 10.1182/blood-2018-04-845834] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/04/2018] [Indexed: 12/12/2022] Open
Abstract
CD137 and its ligand, CD137L, are expressed on activated T cells and antigen-presenting cells, respectively. Recent studies have shown that CD137L and CD137 are aberrantly expressed by tumor cells, especially in some hematopoietic malignancies, and interactions between these molecules on tumor cells promote tumor growth. In this study, we investigated the roles of CD137L and CD137 in cutaneous T-cell lymphoma (CTCL), represented by mycosis fungoides and Sézary syndrome. Flow cytometric analysis showed that primary Sézary cells and CTCL cell lines (Hut78, MyLa, HH, SeAx, and MJ) aberrantly expressed CD137L. CD137L expression by tumor cells in CTCL was also confirmed by immunohistochemistry. Anti-CD137L-neutralizing antibody inhibited proliferation, survival, CXCR4-mediated migration, and in vivo growth in CTCL cell lines through inhibition of phosphorylation of AKT, extracellular signal-regulated kinase 1/2, p38 MAPK, and JNK. Moreover, suppression of CD137L signaling decreased antiapoptotic proteins Bcl-2 and phosphorylated Bad. We also explored the transcription factor regulating CD137L expression. Because GATA6 has been proposed as an oncogene in many types of tumors with aberrant CD137L expression, we examined GATA6 expression and the involvement of GATA6 in CD137L expression in CTCL. DNA hypomethylation and histone acetylation induced GATA6 overexpression in CTCL cells. Furthermore, chromatin immunoprecipitation, luciferase reporter assay, and knockdown by short hairpin RNA showed that GATA6 directly upregulated CD137L expression. Inhibition of GATA6 resulted in decreased survival and in vivo growth in CTCL cells. Collectively, our findings prompt a novel therapeutic approach to CTCL based on the discovery that the GATA6/CD137L axis plays an important role in the tumorigenesis of CTCL.
Collapse
|
22
|
Panjwani PK, Charu V, DeLisser M, Molina-Kirsch H, Natkunam Y, Zhao S. Programmed death-1 ligands PD-L1 and PD-L2 show distinctive and restricted patterns of expression in lymphoma subtypes. Hum Pathol 2018; 71:91-99. [DOI: 10.1016/j.humpath.2017.10.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/22/2017] [Accepted: 10/25/2017] [Indexed: 12/13/2022]
|
23
|
Activation of CD137 signaling accelerates vascular calcification in vivo and vitro. Int J Cardiol 2017; 230:198-203. [DOI: 10.1016/j.ijcard.2016.12.174] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/18/2016] [Accepted: 12/25/2016] [Indexed: 01/08/2023]
|
24
|
Shi YF, Gao ZF, Liu CL, Huang X, Song YQ, Zhang C, Lin DM, Zhou LX, Zhao M, Lai YM, Li M. [Expression of CD137 in tumor cells of Hodgkin lymphoma from Northern China and its application in pathological differential diagnosis]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2017; 37:484-90. [PMID: 27431073 PMCID: PMC7348333 DOI: 10.3760/cma.j.issn.0253-2727.2016.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
目的 明确CD137在北方地区经典型霍奇金淋巴瘤(cHL)中的表达,探讨其作为cHL辅助病理鉴别诊断新指标的可能应用价值。 方法 收集54例cHL患者资料,以55例伴有“HRS样细胞”的非cHL患者为对照。在病理组织标本中选取“HRS细胞”或“HRS样细胞”丰富的区域制作组织芯片;以“HRS细胞”或“HRS样细胞”为观察对象,cHL组应用CD30、CD15、CD20、PAX5、CD3免疫组织化学染色;同时对两组患者标本进行CD137(BBK-2)抗体免疫组织化学染色及采用EBV编码的小RNA(EBER)原位杂交法检测EBV感染状态。 结果 54例cHL患者均为淋巴结内原发,中位年龄45.5(22.0~68.0)岁;男女比例1.7∶1;对照组患者结内54例,结外(皮肤)1例,中位年龄50.0(12.0~81.0)岁;男女比例1.9∶1。54例cHL患者均表达CD30,HRS细胞主要诊断相关免疫标志物CD30、CD15、CD20、CD3阳性表达率依次为100.0%、70.4%、18.5%和0,可见PAX5弱至中等强度表达,阳性率70.4%;EBV感染阳性率25.9%(对照组阳性率21.8%)。cHL组CD137阳性率57.4%,对照组阳性率14.5%,差异有统计学意义(P<0.001)。将cHL组及对照组按照患者年龄(≥60/<60岁)、性别、有无EBV感染、组织学亚型以及主要诊断相关标志物的表达与否进行分组,CD137阳性率差异均无统计学意义(P值均> 0.05)。以2013年为界进行分组,2013年前后两组cHL患者的CD137阳性率差异有统计学意义(39.4%对85.7%,P=0.001),对照组差异无统计学意义(12.5%对16.1%,P=0.705);2013以后存档的标本中cHL组与对照组患者CD137阳性率差异有统计学意义(85.7%对16.1%,P<0.001)。 结论 通过研究初步证实北方地区大多数cHL患者的HRS细胞表达CD137,而对照组患者“HRS样细胞”CD137阳性率较低。保存期3年以内较保存期3年以上的cHL患者标本CD137阳性率高,更适于进行CD137免疫组织化学染色检测。CD137有望作为辅助cHL病理鉴别诊断的新指标。
Collapse
Affiliation(s)
- Y F Shi
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | | | | | | | | | | | | | | | | | | | - M Li
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
25
|
Chester C, Ambulkar S, Kohrt HE. 4-1BB agonism: adding the accelerator to cancer immunotherapy. Cancer Immunol Immunother 2016; 65:1243-8. [PMID: 27034234 PMCID: PMC5035667 DOI: 10.1007/s00262-016-1829-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/21/2016] [Indexed: 01/06/2023]
Abstract
The success of checkpoint inhibitors has validated immunomodulatory agents as a valuable class of anticancer therapeutics. A promising co-stimulatory immunologic target is 4-1BB, or CD137, a member of the tumor necrosis factor receptor superfamily. Ligation of 4-1BB induces an activating signal in CD8(+) T cells and natural killer cells, resulting in increased pro-inflammatory cytokine secretion, cytolytic function, and antibody-dependent cell-mediated cytotoxicity. Targeting 4-1BB with agonistic monoclonal antibody (mAb) therapy demonstrated potent antitumor effects in murine tumor models. While anti-4-1BB mAbs have entered clinical trials, optimal efficacy of 4-1BB-targeted agents will inevitably come from combination therapeutic strategies. Checkpoint blockade is a compelling combination partner for 4-1BB agonism. This novel immunotherapeutic approach has the potential to active antitumor immune effectors by a complementary mechanism: simultaneously "removing the brakes" via blocking inhibitory signaling and "stepping on the accelerator" via co-stimulation. While important considerations should be given to 4-1BB-mediated toxicities, the current understanding of 4-1BB biology suggests it may play a key role in advancing the capabilities of cancer combination therapy.
Collapse
Affiliation(s)
- Cariad Chester
- Department of Medicine, Division of Oncology, Stanford University Medical Center, Stanford University, 269 Campus Drive, CCSR 1140, Stanford, CA, 94305-5151, USA.
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Siddhant Ambulkar
- Department of Medicine, Division of Oncology, Stanford University Medical Center, Stanford University, 269 Campus Drive, CCSR 1140, Stanford, CA, 94305-5151, USA
| | - Holbrook E Kohrt
- Department of Medicine, Division of Oncology, Stanford University Medical Center, Stanford University, 269 Campus Drive, CCSR 1140, Stanford, CA, 94305-5151, USA
| |
Collapse
|
26
|
Integrated mate-pair and RNA sequencing identifies novel, targetable gene fusions in peripheral T-cell lymphoma. Blood 2016; 128:1234-45. [PMID: 27297792 DOI: 10.1182/blood-2016-03-707141] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/25/2016] [Indexed: 12/15/2022] Open
Abstract
Peripheral T-cell lymphomas (PTCLs) represent a heterogeneous group of T-cell malignancies that generally demonstrate aggressive clinical behavior, often are refractory to standard therapy, and remain significantly understudied. The most common World Health Organization subtype is PTCL, not otherwise specified (NOS), essentially a "wastebasket" category because of inadequate understanding to assign cases to a more specific diagnostic entity. Identification of novel fusion genes has contributed significantly to improving the classification, biologic understanding, and therapeutic targeting of PTCLs. Here, we integrated mate-pair DNA and RNA next-generation sequencing to identify chromosomal rearrangements encoding expressed fusion transcripts in PTCL, NOS. Two of 11 cases had novel fusions involving VAV1, encoding a truncated form of the VAV1 guanine nucleotide exchange factor important in T-cell receptor signaling. Fluorescence in situ hybridization studies identified VAV1 rearrangements in 10 of 148 PTCLs (7%). These were observed exclusively in PTCL, NOS (11%) and anaplastic large cell lymphoma (11%). In vitro, ectopic expression of a VAV1 fusion promoted cell growth and migration in a RAC1-dependent manner. This growth was inhibited by azathioprine, a clinically available RAC1 inhibitor. We also identified novel kinase gene fusions, ITK-FER and IKZF2-ERBB4, as candidate therapeutic targets that show similarities to known recurrent oncogenic ITK-SYK fusions and ERBB4 transcript variants in PTCLs, respectively. Additional novel and potentially clinically relevant fusions also were discovered. Together, these findings identify VAV1 fusions as recurrent and targetable events in PTCLs and highlight the potential for clinical sequencing to guide individualized therapy approaches for this group of aggressive malignancies.
Collapse
|
27
|
Rajendran S, Ho WT, Schwarz H. CD137 signaling in Hodgkin and Reed-Sternberg cell lines induces IL-13 secretion, immune deviation and enhanced growth. Oncoimmunology 2016; 5:e1160188. [PMID: 27471634 PMCID: PMC4938358 DOI: 10.1080/2162402x.2016.1160188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/13/2016] [Accepted: 02/25/2016] [Indexed: 01/05/2023] Open
Abstract
CD137 and its ligand, CD137L, are expressed on activated T cells and antigen-presenting cells (APC), respectively, and are powerful inducers of cellular, type 1 immune responses. CD137 is ectopically expressed by Hodgkin and Reed-Sternberg (HRS) cells, the malignant cells in Hodgkin lymphoma (HL). Here we report that CD137 transmits signals into HRS cells, which induce the secretion of IL-13. IL-13 in conditioned supernatants of HRS cell lines inhibits the secretion of IFNγ by peripheral blood mononuclear cells (PBMC). Since IFNγ is essential for the development of a type 1 immune response, CD137-induced IL-13 secretion facilitates escape from immune surveillance. Further, CD137-induced IL-13 enhances the growth of HRS cell lines. CD137, IL-13 double-positive cells could be detected in the majority (58%) of HL patient samples, providing clinical evidence for a role of IL-13 induction by CD137 during HL pathogenesis. This study validates CD137 as a candidate target for immunotherapy of HL.
Collapse
Affiliation(s)
| | | | - Herbert Schwarz
- Department of Physiology
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| |
Collapse
|
28
|
Verma A, Shet T, Epari S, Gupta M, Gujral S, Khanna N, Laskar S, Sengar M, Arora B, Menon H, Banavali S. Mediastinal Gray Zone Lymphoma. Int J Surg Pathol 2016; 24:382-93. [DOI: 10.1177/1066896916635816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Aim. To identify aggressively behaving classical Hodgkin lymphoma (CHL) of mediastinum and primary mediastinal B-cell lymphoma (PMBCL) and to classify them as mediastinal gray zone lymphoma(MGZL) and to define a minimum immunopanel for the diagnosis of MGZL. Materials and Methods. Ninety-two mediastinal B-cell lymphomas were reviewed with a wide immunopanel and were classified as CHL, PMBCL, or MGZL. CHL with an expression of 3 or 4 transcription factors performed worse, and hence the CHL with ≥3 transcription factors were classified as MGZL-CHL. In PMBCL, the cases with a weak or negative CD20 and positive CD15 as well as those cases showing cyclin E positivity with a negative or focal LCA and any one of the transcription factors were classified as MGZL-PMBCL. Results. The MGZL cases expanded from 9 to 28 cases after using an extended immunopanel. CHL and PMBCL had a disease-free survival rate of 86.8% and 69.2% and an overall survival rate of 97.4% and 80.8%, respectively. MGZL-CHL and MGZL-PMBCL had a disease-free survival rate of 33% and 40% and an overall survival rate of 66.7% and 60%, respectively. Conclusion. Thus, the MGZL may be a wider category than we think and hence the use of a wide immunopanel is recommended to identify the aggressively behaving mediastinal B-cell lymphomas.
Collapse
Affiliation(s)
- Anuj Verma
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Tanuja Shet
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Sridhar Epari
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Manjudevi Gupta
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Sumeet Gujral
- Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - Nehal Khanna
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, India
| | - Siddhartha Laskar
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, India
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Brijesh Arora
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, India
| | - Hari Menon
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Shripad Banavali
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
29
|
Makkouk A, Chester C, Kohrt HE. Rationale for anti-CD137 cancer immunotherapy. Eur J Cancer 2016; 54:112-119. [PMID: 26751393 DOI: 10.1016/j.ejca.2015.09.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 09/21/2015] [Accepted: 09/21/2015] [Indexed: 11/29/2022]
Abstract
The consideration of the complex interplay between the tumour microenvironment (TME) and the immune response is the key for designing effective immunotherapies. Therapeutic strategies that harness co-stimulatory receptors have recently gained momentum in the clinic. One such strategy with promising clinical applications is the targeting of CD137, a member of the tumour necrosis factor receptor superfamily. Its expression on both innate and adaptive immune cells, coupled with its unique ability to potentiate antitumour responses through modulating the TME and to ameliorate autoimmune responses, has established it as an appealing target. In this review, we will discuss the various CD137-targeted immunotherapeutics that have reached clinical development, with a focus on recent advances and novel modalities such as CD137 chimeric antigen receptors and CD137 bispecific antibodies. We will also highlight the effect of CD137 targeting on the TME and discuss the importance of probing TME changes for predicting and testing the efficacy of CD137-mediated immunotherapy.
Collapse
Affiliation(s)
- Amani Makkouk
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305 USA
| | - Cariad Chester
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305 USA; Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Holbrook E Kohrt
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA 94305 USA.
| |
Collapse
|
30
|
Wilcox RA. A three-signal model of T-cell lymphoma pathogenesis. Am J Hematol 2016; 91:113-22. [PMID: 26408334 DOI: 10.1002/ajh.24203] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 08/24/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022]
Abstract
T-cell lymphoma pathogenesis and classification have, until recently, remained enigmatic. Recently performed whole-exome sequencing and gene-expression profiling studies have significant implications for their classification and treatment. Recurrent genetic modifications in antigen ("signal 1"), costimulatory ("signal 2"), or cytokine receptors ("signal 3"), and the tyrosine kinases and other signaling proteins they activate, have emerged as important therapeutic targets in these lymphomas. Many of these genetic modifications do not function in a cell-autonomous manner, but require the provision of ligand(s) by constituents of the tumor microenvironment, further supporting the long-appreciated view that these lymphomas are dependent upon and driven by their microenvironment. Therefore, the seemingly disparate fields of genomics and immunology are converging. A unifying "3 signal model" for T-cell lymphoma pathogenesis that integrates these findings will be presented, and its therapeutic implications briefly reviewed.
Collapse
Affiliation(s)
- Ryan A. Wilcox
- Department of Internal Medicine, Division of Hematology and Oncology; University of Michigan; Ann Arbor Michigan
| |
Collapse
|
31
|
Bartkowiak T, Curran MA. 4-1BB Agonists: Multi-Potent Potentiators of Tumor Immunity. Front Oncol 2015; 5:117. [PMID: 26106583 PMCID: PMC4459101 DOI: 10.3389/fonc.2015.00117] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/11/2015] [Indexed: 01/12/2023] Open
Abstract
Immunotherapy is a rapidly expanding field of oncology aimed at targeting, not the tumor itself, but the immune system combating the cancerous lesion. Of the many approaches currently under study to boost anti-tumor immune responses; modulation of immune co-receptors on lymphocytes in the tumor microenvironment has thus far proven to be the most effective. Antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte antigen-4 (CTLA-4) has become the first FDA approved immune checkpoint blockade; however, tumor infiltrating lymphocytes express a diverse array of additional stimulatory and inhibitory co-receptors, which can be targeted to boost tumor immunity. Among these, the co-stimulatory receptor 4-1BB (CD137/TNFSF9) possesses an unequaled capacity for both activation and pro-inflammatory polarization of anti-tumor lymphocytes. While functional studies of 4-1BB have focused on its prominent role in augmenting cytotoxic CD8 T cells, 4-1BB can also modulate the activity of CD4 T cells, B cells, natural killer cells, monocytes, macrophages, and dendritic cells. 4-1BB’s expression on both T cells and antigen presenting cells, coupled with its capacity to promote survival, expansion, and enhanced effector function of activated T cells, has made it an alluring target for tumor immunotherapy. In contrast to immune checkpoint blocking antibodies, 4-1BB agonists can both potentiate anti-tumor and anti-viral immunity, while at the same time ameliorating autoimmune disease. Despite this, 4-1BB agonists can trigger high grade liver inflammation which has slowed their clinical development. In this review, we discuss how the underlying immunobiology of 4-1BB activation suggests the potential for therapeutically synergistic combination strategies in which immune adverse events can be minimized.
Collapse
Affiliation(s)
- Todd Bartkowiak
- Department of Immunology, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences at Houston , Houston, TX , USA
| | - Michael A Curran
- Department of Immunology, University of Texas MD Anderson Cancer Center , Houston, TX , USA ; The University of Texas Graduate School of Biomedical Sciences at Houston , Houston, TX , USA
| |
Collapse
|
32
|
Shao Z, Harfuddin Z, Pang WL, Nickles E, Koh LK, Schwarz H. Trogocytic CD137 transfer causes an internalization of CD137 ligand on murine APCs leading to reduced T cell costimulation. J Leukoc Biol 2015; 97:909-919. [DOI: 10.1189/jlb.3a0213-079rrr] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Abstract
CD137 ligand (CD137L) is expressed on APCs and crosslinks CD137, a powerful costimulatory molecule on T cells during cognate interactions, and thereby greatly enhances immune responses. We report that CD137 can be transferred from activated T cells and from tumor cells that express CD137 to other cells via trogocytosis. This trogocytic transfer is independent of CD137L expression by the recipient cell. However, if CD137L is present on the recipient cell, the transferred CD137 binds to CD137L and the CD137-CD137L complex becomes internalized. The removal of CD137L from the surface of APCs lowers their ability to costimulate T cells, as evidenced by a reduced IFN-γ secretion. Removal of CD137L on APCs by trogocytic transfer of CD137 occurs within 1 h and requires cell-cell contact and the continuous presence of CD137-expressing cells. Bidirectional signaling exists for the CD137 receptor/ligand system, because CD137L also signals into APCs. We propose that the trogocytic transfer of CD137 from activated T cells to APCs and the subsequent removal of CD137L from APCs is a physiologic regulatory mechanism that limits immune activity. Furthermore, we hypothesize that the trogocytic transfer of CD137 occurs in cancers and quenches the activity of APCs, contributing to the cancer cells escaping immune surveillance. Taken together, our findings demonstrate that the trogocytic transfer of CD137 leads to an internalization of CD137L on APCs and a reduction in immune activity.
Collapse
Affiliation(s)
- Zhe Shao
- Department of Physiology, National University of Singapore , Singapore , Singapore
- Immunology Programme, National University of Singapore , Singapore , Singapore
| | - Zulkarnain Harfuddin
- Department of Physiology, National University of Singapore , Singapore , Singapore
- Immunology Programme, National University of Singapore , Singapore , Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore , Singapore
| | - Wan Lu Pang
- Department of Physiology, National University of Singapore , Singapore , Singapore
- Immunology Programme, National University of Singapore , Singapore , Singapore
| | - Emily Nickles
- Department of Physiology, National University of Singapore , Singapore , Singapore
- Immunology Programme, National University of Singapore , Singapore , Singapore
| | - Liang Kai Koh
- Department of Physiology, National University of Singapore , Singapore , Singapore
- Immunology Programme, National University of Singapore , Singapore , Singapore
| | - Herbert Schwarz
- Department of Physiology, National University of Singapore , Singapore , Singapore
- Immunology Programme, National University of Singapore , Singapore , Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore , Singapore , Singapore
| |
Collapse
|
33
|
Vinay DS, Kwon BS. 4-1BB (CD137), an inducible costimulatory receptor, as a specific target for cancer therapy. BMB Rep 2014; 47:122-9. [PMID: 24499671 PMCID: PMC4163883 DOI: 10.5483/bmbrep.2014.47.3.283] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 12/30/2013] [Accepted: 01/28/2014] [Indexed: 01/09/2023] Open
Abstract
Although considerable progress has been made in understanding how tumors evade immune surveillance, measures to counter the same have not kept pace with the advances made in designing effective strategies. 4-1BB (CD137; TNFRS9), an activation-induced costimulatory molecule, is an important regulator of immune responses. Targeting 4-1BB or its natural ligand 4-1BB ligand (4-1BBL) has important implications in many clinical conditions, including cancer. In-depth analysis revealed that 4-1BB-mediated anti-cancer effects are based on its ability to induce activation of cytotoxic T lymphocytes (CTL), and among others, high amounts of IFN-γ. In this review, we will discuss the various aspects of 4-1BB-mediated anti-tumor responses, the basis of such responses, and future directions. [BMB Reports 2014; 47(3): 122-129]
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA
| | - Byoung S Kwon
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA70112, USA; Cell and Immunobiology, and R & D Center for Cancer Therapeutics, National Cancer Center, Goyang 410-769, Korea
| |
Collapse
|
34
|
Zhang X(M, Aguilera N. New Immunohistochemistry for B-Cell Lymphoma and Hodgkin Lymphoma. Arch Pathol Lab Med 2014; 138:1666-72. [DOI: 10.5858/arpa.2014-0058-ra] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context
B-cell non-Hodgkin lymphoma is a heterogeneous group of lymphoproliferative malignancies with different clinical behaviors and treatments. It is important to differentiate individual B-cell lymphoma to apply the best treatment and management. Morphology and immunohistochemistry are the primary tools used for diagnosing lymphoma. There is a characteristic pattern of expression with immunohistochemical antibodies in most well-defined B-cell lymphomas. Some cases of B-cell lymphoma, however, show unusual morphologic and immunophenotypic features. The new and sometimes more specific antibodies have been developed recently, which may further define those lymphomas. Only with use of the antibodies over time does their true nature and specificity become evident.
Objectives
To present new antibodies for B-cell lymphoma that enhance the probability for diagnosis or can act as alternate markers in unusual cases, in which a B-cell lymphoma does not present with characteristic immunohistochemical staining, and to present prognostic markers that allow for better management of patients with specific B-cell lymphomas.
Data Sources
Data were obtained from literature review and figures from slides in personal practice.
Conclusions
The immunohistochemical antibodies presented in this article increase our ability to understand, diagnosis, and manage patients with B-cell lymphoma.
Collapse
Affiliation(s)
- Xiaohong (Mary) Zhang
- From the Department of Laboratory Medicine, Geisinger Medical Laboratories, Wilkes-Barre, Pennsylvania (Dr Zhang)
| | - Nadine Aguilera
- and the Department of Pathology, University of Virginia Health System, Charlottesville (Dr Aguilera)
| |
Collapse
|
35
|
CD137 expression is induced by Epstein-Barr virus infection through LMP1 in T or NK cells and mediates survival promoting signals. PLoS One 2014; 9:e112564. [PMID: 25409517 PMCID: PMC4237363 DOI: 10.1371/journal.pone.0112564] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 10/20/2014] [Indexed: 12/15/2022] Open
Abstract
To clarify the mechanism for development of Epstein-Barr virus (EBV)-positive T- or NK-cell neoplasms, we focused on the costimulatory receptor CD137. We detected high expression of CD137 gene and its protein on EBV-positive T- or NK-cell lines as compared with EBV-negative cell lines. EBV-positive cells from EBV-positive T- or NK-cell lymphoproliferative disorders (EBV-T/NK-LPDs) patients also had significantly higher CD137 gene expression than control cells from healthy donors. In the presence of IL-2, whose concentration in the serum of EBV-T/NK-LPDs was higher than that of healthy donors, CD137 protein expression was upregulated in the patients' cells whereas not in control cells from healthy donors. In vitro EBV infection of MOLT4 cells resulted in induction of endogenous CD137 expression. Transient expression of LMP1, which was enhanced by IL-2 in EBV-T/NK-LPDs cells, induced endogenous CD137 gene expression in T and NK-cell lines. In order to examine in vivo CD137 expression, we used EBV-T/NK-LPDs xenograft models generated by intravenous injection of patients' cells. We identified EBV-positive and CD8-positive T cells, as well as CD137 ligand-positive cells, in their tissue lesions. In addition, we detected CD137 expression on the EBV infected cells from the lesions of the models by immune-fluorescent staining. Finally, CD137 stimulation suppressed etoposide-induced cell death not only in the EBV-positive T- or NK-cell lines, but also in the patients' cells. These results indicate that upregulation of CD137 expression through LMP1 by EBV promotes cell survival in T or NK cells leading to development of EBV-positive T/NK-cell neoplasms.
Collapse
|
36
|
Zhao S, Xing Y, Natkunam Y. Use of CD137 ligand expression in the detection of small B-cell lymphomas involving the bone marrow. Hum Pathol 2014; 45:1024-30. [PMID: 24746207 DOI: 10.1016/j.humpath.2013.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/20/2013] [Accepted: 12/27/2013] [Indexed: 10/25/2022]
Abstract
Staging for small B-cell lymphomas is important for prognostic and therapeutic decision making; however, the detection of lymphoid infiltrates in the bone marrow is often hampered by the lack of specific diagnostic markers. We recently described the hematopoietic tissue distribution patterns of CD137 and CD137 ligand (CD137L), which have shown promise as immunotherapeutic targets. CD137 expression was primarily confined to cells in the microenvironment, whereas CD137L was expressed in neoplastic cells in most B-cell lymphomas. Here we evaluate the use of CD137L in the detection of small B-cell lymphomas involving the bone marrow. To test the potential efficacy of CD137L in detecting bone marrow lymphoid infiltrates, 166 small B-cell lymphomas were evaluated by immunohistochemistry and double-immunofluorescence labeling on formalin-fixed, paraffin-embedded bone marrow core biopsies. CD137L was highly expressed in bone marrows involved by small B-cell lymphomas and included hairy cell leukemia, mantle cell lymphoma, follicular lymphoma, B-lymphoblastic leukemia, and chronic lymphocytic leukemia. In addition, a small subset of marginal zone lymphoma and most of lymphoplasmacytic lymphoma showed staining. Normal bone marrow cells including myeloid, erythroid and megakaryocytic precursors, and reactive lymphoid aggregates lacked staining. Our findings show that immunohistochemistry for CD137L is capable of reliably distinguishing small B-cell lymphomas from reactive lymphoid aggregates. These data also suggest that CD137L is useful in providing staging information for clinical diagnosis and is likely to furnish a potential target for minimal residual disease assessment as well as immunotherapy in patients with stage 4 disease.
Collapse
Affiliation(s)
- Shuchun Zhao
- Stem Cell Center, Zhengzhou University School of Medicine, Zhengzhou 450001, China; Department of Pathology, Stanford University School of Medicine, Stanford 94305, CA
| | - Ying Xing
- Stem Cell Center, Zhengzhou University School of Medicine, Zhengzhou 450001, China.
| | - Yasodha Natkunam
- Department of Pathology, Stanford University School of Medicine, Stanford 94305, CA.
| |
Collapse
|
37
|
He Y, Wang J, Gou L, Shen C, Chen L, Yi C, Wei X, Yang J. Comprehensive analysis of expression profile reveals the ubiquitous distribution of PPPDE peptidase domain 1, a Golgi apparatus component, and its implications in clinical cancer. Biochimie 2013; 95:1466-75. [PMID: 23567336 DOI: 10.1016/j.biochi.2013.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 03/26/2013] [Indexed: 02/05/2023]
Abstract
PPPDE peptidase domain 1 (PPPDE1) is a recently identified gene; however, its expression regulation and biological function are unclear. Previous studies have indicated that PPPDE1 is involved in embryogenesis, apoptosis induction and cell cycle regulation. In the present study, we first used an anti-PPPDE1 antibody to determine that endogenous PPPDE1 is located in the Golgi apparatus. Immunohistochemistry (IHC) of mouse embryos indicated that PPPDE1 was markedly distributed in liver, skin, intestinal villi, and muscles, whereas Western blot analysis of mouse mature organs revealed its ubiquitous expression, without an appreciable distinction in protein abundance. Surprisingly, another potential isoform of PPPDE1 with a molecular weight of 18 kD (rather than its predicted molecular weight of 21 kD) was detected in the mouse kidney, testis, and intestine. Moreover, microarrays that were derived from twelve tumor types revealed that PPPDE1 expression was significantly lower in pancreas, stomach, and skin tumors compared with normal tissue from these organs. We specifically and extensively analyzed PPPDE1 expression in clinical samples and observed strong associations between PPPDE1 expression and (i) differentiation grade in pancreatic ductal adenocarcinoma and (ii) T stage in skin squamous cell carcinoma. Our data are the first to reveal the expression profile of PPPDE1 protein and its implications in cancer. These results will contribute to the understanding of the expression regulation and biological functions of PPPDE1 in development and carcinogenesis.
Collapse
Affiliation(s)
- Yi He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, No. 1, Keyuan Road 4, Gaopeng Street, Chengdu 610041, China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Pang WL, Ho WT, Schwarz H. Ectopic CD137 expression facilitates the escape of Hodgkin and Reed-Sternberg cells from immunosurveillance. Oncoimmunology 2013; 2:e23441. [PMID: 23734307 PMCID: PMC3654577 DOI: 10.4161/onci.23441] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 12/29/2012] [Indexed: 11/19/2022] Open
Abstract
CD137 is ectopically expressed on Hodgkin and Reed-Sternberg (HRS) cells, causing the removal of the immunostimulatory CD137 ligand from HRS cells as well as from surrounding antigen presenting cells. This inhibits T-cell co-stimulation and supports the immune evasion of Hodgkin's lymphoma.
Collapse
Affiliation(s)
- Wan Lu Pang
- Department of Physiology and Immunology Programme; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Weng Tong Ho
- Department of Physiology and Immunology Programme; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
| | - Herbert Schwarz
- Department of Physiology and Immunology Programme; Yong Loo Lin School of Medicine; National University of Singapore; Singapore
- NUS Graduate School for Integrative Sciences and Engineering; National University of Singapore; Singapore
| |
Collapse
|
39
|
Moran AE, Kovacsovics-Bankowski M, Weinberg AD. The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy. Curr Opin Immunol 2013; 25:230-7. [PMID: 23414607 DOI: 10.1016/j.coi.2013.01.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 01/15/2013] [Indexed: 12/23/2022]
Abstract
T cell-mediated rejection of tumors requires signals from the T cell receptor and co-stimulatory molecules to license effector functions of tumor-antigen specific T cells. There is also an array of immune suppressive mechanisms within the tumor microenvironment that can suppress anti-tumor immunity. The use of monoclonal antibodies to overcome this suppression and/or enhance tumor-antigen specific T cell responses has shown promise in clinical trials. In particular, targeting co-stimulatory members of the tumor necrosis factor receptor (TNFR) family with agonist Abs enhances T cell function, which has led to encouraging therapeutic results in cancer-bearing hosts. These encouraging data establish TNFRs as important targets for enhancing tumor-specific immune responses in mice and man. This review will focus on agonists that target the TNFRs OX40, 4-1BB, and CD40.
Collapse
Affiliation(s)
- Amy E Moran
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Research Center, Providence Portland Medical Center, 4805 NE Glisan St., Portland, OR 97213, USA
| | | | | |
Collapse
|
40
|
Ho WT, Pang WL, Chong SM, Castella A, Al-Salam S, Tan TE, Moh MC, Koh LK, Gan SU, Cheng CK, Schwarz H. Expression of CD137 on Hodgkin and Reed–Sternberg Cells Inhibits T-cell Activation by Eliminating CD137 Ligand Expression. Cancer Res 2012. [DOI: 10.1158/0008-5472.can-12-3849] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|