51
|
Lenouvel D, González-Moles MÁ, Talbaoui A, Ramos-García P, González-Ruiz L, Ruiz-Ávila I, Gil-Montoya JA. An update of knowledge on PD-L1 in head and neck cancers: Physiologic, prognostic and therapeutic perspectives. Oral Dis 2019; 26:511-526. [PMID: 30866171 DOI: 10.1111/odi.13088] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/09/2019] [Accepted: 03/07/2019] [Indexed: 12/24/2022]
Abstract
Programmed cell death-ligand 1 (PD-L1) is a transmembrane protein that acts as a co-inhibitory factor in the immune response. Its receptor, programmed cell death protein 1 (PD-1), is found on immune cells, where binding to PD-L1 can reduce the proliferation of PD-1-positive cells, inhibit their cytokine secretion and induce apoptosis. PD-L1 in immune-privileged tissue plays a crucial role in peripheral tolerance. PD-L1 can be overexpressed in various malignancies, including oral squamous cell carcinoma, where it can attenuate the host immune response to tumour cells and has been associated with a worse prognosis. Monoclonal antibody therapies targeting the PD-1:PD-L1 axis have shown initial promise, but further research is needed to identify which patients will benefit. We provide an update of knowledge on PD-L1, including its structure, function and regulation. We also review studies on the overexpression of PD-L1 in cancer, specifically oral squamous cell carcinoma, and explore its potential value as a therapeutic target.
Collapse
Affiliation(s)
| | - Miguel Ángel González-Moles
- School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, Granada, Spain
| | - Asmae Talbaoui
- School of Dentistry, University of Granada, Granada, Spain
| | | | - Lucía González-Ruiz
- Servicio de Dermatología, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Isabel Ruiz-Ávila
- Instituto de Investigación Biosanitaria, Granada, Spain.,Servicio de Anatomía Patológica, Complejo Hospitalario Universitario de Granada, Granada, Spain
| | - José Antonio Gil-Montoya
- School of Dentistry, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria, Granada, Spain
| |
Collapse
|
52
|
Bang YJ, Van Cutsem E, Fuchs CS, Ohtsu A, Tabernero J, Ilson DH, Hyung WJ, Strong VE, Goetze TO, Yoshikawa T, Tang LH, Hwang PMT, Webb N, Adelberg D, Shitara K. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol 2019; 15:943-952. [PMID: 30777447 DOI: 10.2217/fon-2018-0581] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Surgical resection is the only curative treatment option for gastric cancer. Despite widespread adoption of multimodality perioperative treatment strategies, 5-year overall survival rates remain low. In patients with advanced gastric or gastroesophageal junction adenocarcinoma, pembrolizumab has demonstrated promising efficacy and manageable safety as monotherapy in previously treated patients and as first-line therapy in combination with cisplatin and 5-fluorouracil. Combining chemotherapy with pembrolizumab in the neoadjuvant/adjuvant setting may benefit patients with locally advanced, resectable disease. AIM To describe the design and rationale for the global, multicenter, randomized, double-blind, Phase III KEYNOTE-585 study to evaluate the efficacy and safety of pembrolizumab plus chemotherapy compared with placebo plus chemotherapy as neoadjuvant/adjuvant treatment for localized gastric or gastroesophageal junction adenocarcinoma. ClinicalTrials.gov : NCT03221426.
Collapse
Affiliation(s)
- Yung-Jue Bang
- Seoul National University College of Medicine, 101 Daehak-ro, Ihwa-Dong, Seoul, Republic of Korea
| | - Eric Van Cutsem
- University Hospitals Gasthuisberg Leuven & KU Leuven, 49 Herestraat, Leuven 3001, Belgium
| | - Charles S Fuchs
- Yale Cancer Center and Smilow Cancer Hospital, 333 Cedar Street, New Haven, CT 06510, USA
| | - Atsushi Ohtsu
- National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | - Josep Tabernero
- Vall d'Hebron University Hospital & Institute of Oncology (VHIO), Vall d'Hebron 119-129, 08035 Universitat Autònoma de Barcelona, Barcelona, Spain
| | - David H Ilson
- Memorial Sloan Kettering Cancer Center, 300 East 66th Street, BAIC 1031, New York, NY 10065, USA
| | - Woo Jin Hyung
- Yonsei Cancer Hospital, Yonsei University Health System, 50-1 Yonsei-Ro Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Vivian E Strong
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Thorsten Oliver Goetze
- Institute of Clinical Cancer Research, UCT University Cancer Center Frankfurt, Steinbacher Hohl 2-26, Frankfurt 60488, Germany
| | - Takaki Yoshikawa
- National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Laura H Tang
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | | | - Nancy Webb
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - David Adelberg
- Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ 07033, USA
| | - Kohei Shitara
- National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| |
Collapse
|
53
|
Reyes VE, Peniche AG. Helicobacter pylori Deregulates T and B Cell Signaling to Trigger Immune Evasion. Curr Top Microbiol Immunol 2019; 421:229-265. [PMID: 31123892 DOI: 10.1007/978-3-030-15138-6_10] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori is a prevalent human pathogen that successfully establishes chronic infection, which leads to clinically significant gastric diseases including chronic gastritis, peptic ulcer disease (PUD), and gastric cancer (GC). H. pylori is able to produce a persistent infection due in large part to its ability to hijack the host immune response. The host adaptive immune response is activated to strategically and specifically attack pathogens and normally clears them from the infected host. Since B and T lymphocytes are central mediators of adaptive immunity, in this chapter we review their development and the fundamental mechanisms regulating their activation in order to understand how some of the normal processes are subverted by H. pylori. In this review, we place particular emphasis on the CD4+ T cell responses, their subtypes, and regulatory mechanisms because of the expanding literature in this area related to H. pylori. T lymphocyte differentiation and function are finely orchestrated through a series of cell-cell interactions, which include immune checkpoint receptors. Among the immune checkpoint receptor family, there are some with inhibitory properties that are exploited by tumor cells to facilitate their immune evasion. Gastric epithelial cells (GECs), which act as antigen-presenting cells (APCs) in the gastric mucosa, are induced by H. pylori to express immune checkpoint receptors known to sway T lymphocyte function and thus circumvent effective T effector lymphocyte responses. This chapter reviews these and other mechanisms used by H. pylori to interfere with host immunity in order to persist.
Collapse
Affiliation(s)
- Victor E Reyes
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| | - Alex G Peniche
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| |
Collapse
|
54
|
Que Y, Fang Z, Guan Y, Xiao W, Xu B, Zhao J, Chen H, Zhang X, Zeng M, Liang Y, Zhang X. LAG-3 expression on tumor-infiltrating T cells in soft tissue sarcoma correlates with poor survival. Cancer Biol Med 2019; 16:331-340. [PMID: 31516753 PMCID: PMC6713642 DOI: 10.20892/j.issn.2095-3941.2018.0306] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objective To elucidate the role and prognostic significance of lymphocyte activation-gene-3 (LAG-3) in soft tissue sarcoma (STS). Methods The expression of LAG-3 in patient and matched normal blood samples was analyzed by flow cytometry. The localization and prognostic values of LAG-3+ cells in 163 STS patients were analyzed by immunohistochemistry. In addition, the expression of tumor-infiltrating CD3+ T, CD4+ T, and CD8+ T cells and their role in the prognosis of STS were evaluated by immunohistochemistry. The effect of LAG-3 blockade was evaluated in an immunocompetent MCA205 fibrosarcoma mouse model.
Results Peripheral CD8+ and CD4+ T cells from STS patients expressed higher levels of LAG-3 than those from healthy donors. LAG-3 expression in STS was significantly associated with a poor clinical outcome (P = 0.038 ) and was correlated with high pathological grade (P < 0.001), advanced tumor stage ( P = 0.016). Additionally, LAG-3 expression was highly correlated with CD8+ T-cell infiltration (r = 0.7034, P < 0.001). LAG-3 was expressed in murine tumor-infiltrating lymphocytes, and its blockade decreased tumor growth and enhanced secretion of interferon-gamma by CD8 + and CD4+ T cells.
Conclusions LAG-3 blockade may be a promising strategy to improve the effects of targeted therapy in STS.
Collapse
Affiliation(s)
- Yi Que
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zhixin Fang
- Department of Laboratory Medicine and Central Laboratories, Guangdong Second Provincial General Hospital, Guangzhou 510000, China
| | - Yuanxiang Guan
- Department of Gastric and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Wei Xiao
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bushu Xu
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jingjing Zhao
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Huoying Chen
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xinke Zhang
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yao Liang
- Department of Gastric and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xing Zhang
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
55
|
Liu XS, Lin XK, Mei Y, Ahmad S, Yan CX, Jin HL, Yu H, Chen C, Lin CZ, Yu JR. Regulatory T Cells Promote Overexpression of Lgr5 on Gastric Cancer Cells via TGF-beta1 and Confer Poor Prognosis in Gastric Cancer. Front Immunol 2019; 10:1741. [PMID: 31417548 PMCID: PMC6682668 DOI: 10.3389/fimmu.2019.01741] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 01/26/2023] Open
Abstract
Background: The leucine-rich repeat containing G protein-coupled receptor 5 (Lgr5) is considered a cancer stem cell marker, and is often overexpressed in tumors. The interaction between Lgr5 and the immune-related tumor microenvironment is not completely understood. The aim of this study was to examine the role of Lgr5 in the microenvironment of gastric cancer (GC), and to explore possible immunological mechanisms influencing Lgr5 expression that are governed by regulatory T cells. Methods: Lgr5 expression was examined in 180 GC tumors by immunohistochemistry, and in 80 pairs of GC tumors for analysis of Th1/Th2 cytokines by ELISA. In addition, SGC7901 cells were co-cultured with patient-derived Tregs, varying concentrations of TGF-β1, TGF-β1 neutralizing antibody, or TGF-β receptor inhibitor SB431542, and Lgr5 and β-catenin expression were examined by qRT-PCR and western blot. Results: In this study, an immunosuppressive microenvironment was associated with high Lgr5 expression in GC. Furthermore, Lgr5 expression was up-regulated in GC cells co-cultured with Tregs or treated with exogenous TGF-β1. This up-regulation was partially inhibited by the TGF-β1 neutralizing antibody, or TGF-β1 receptor antagonist SB431542. β-catenin was up-regulated with high Lgr5 expression induced by exogenous TGF-β1, and this up-regulation was inhibited by SB431542. An increased number of Tregs and high Lgr5 expression in GC tissues were significantly associated with low overall survival. Conclusion: Tregs promoted increased Lgr5 expression in GC cells via TGF-β1 and TGF-β1 signaling pathway, which may involve activation of the Wnt signaling pathway. High Lgr5 expression via TGF-β confer poor prognosis in gastric cancer.
Collapse
Affiliation(s)
- Xiao-Sun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
| | - Xian-Ke Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Mei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sabir Ahmad
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chong-Xian Yan
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Long Jin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cai-Zhao Lin
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ji-Ren Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Ji-Ren Yu
| |
Collapse
|
56
|
Xu S, Cui F, Huang D, Zhang D, Zhu A, Sun X, Cao Y, Ding S, Wang Y, Gao E, Zhang F. PD-L1 monoclonal antibody-conjugated nanoparticles enhance drug delivery level and chemotherapy efficacy in gastric cancer cells. Int J Nanomedicine 2018; 14:17-32. [PMID: 30587982 PMCID: PMC6302817 DOI: 10.2147/ijn.s175340] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background Docetaxel (DOC) is widely used as a chemotherapy drug for various tumors, including gastric cancer (GC), but the clinical application of DOC has been limited due to the hydrophobicity of the drug. We aimed to formulate a multifunctional nanoparticle (NP) system to reduce the side effects of the chemotherapy agent, to promote synergistic therapeutic effects, and to achieve targeted delivery of the therapy. Methods The polyethylene glycol-poly(ε-caprolactone) NPs (PEG-PCL NPs) were prepared by a ring opening copolymerization technique and were then conjugated with a programmed death-ligand 1 (PD-L1) monoclonal antibody (mAb). The effects of the surface coating on particle size, size distribution, zeta potential, drug encapsulation efficiency, loading capacity, and the drug release kinetics were investigated. By using a panel of PD-L1-expressing human GC cell lines and PD-L1-overexpressing cells, we studied cellular uptake, cytotoxic effects, and cellular apoptosis in the presence of PD-L1 mAb-conjugated NPs. Results The characterization of the structure and biological functions of DOC-PEG-PCL-mAb NPs was investigated in vitro. X-ray photoelectron spectroscopy validated the presence of the PD-L1 mAbs on the NP surface. The cellular uptake analysis showed that the antibody-conjugated NPs achieved significantly higher cellular uptake. The results of an in vitro cytotoxicity experiment on three GC lines further proved the targeting effects of the antibody conjugation. In addition, we found that the DOC-PEG-PCL-mAb NPs induced cell apoptosis and enhanced G2-M arrest in cancer cells, indicating the inhibition of microtubule synthesis. When compared with the control groups, DOC-PEG-PCL-mAb NPs are more effective in inhibiting PD-L1 expression in GC cells. Conclusion Our results reported here highlight the biological and clinical potential of DOC-PEG-PCL-mAb NPs using PD-L1 mAbs in GC treatment.
Collapse
Affiliation(s)
- Shijie Xu
- Center for Public Health Research, Medical School, Nanjing University, Nanjing, China
| | - Fangbo Cui
- Department of Oncology, Wannan Medical College, Wuhu, Anhui, China, .,Department of Oncology, The People's Hospital of Ma Anshan, Ma Anshan, Anhui, China,
| | - Dafu Huang
- Department of Oncology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, Jiangsu, China
| | - Dinghu Zhang
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Anqing Zhu
- Department of Oncology, The Affliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu, China
| | - Xia Sun
- Department of Oncology, The Affliated Jiangyin Hospital of Southeast University Medical College, Jiangyin, Jiangsu, China
| | - Yiming Cao
- Department of General Surgery, The People's Hospital of Ma Anshan, Ma Anshan, Anhui, China
| | - Sheng Ding
- Department of General Surgery, The People's Hospital of Ma Anshan, Ma Anshan, Anhui, China
| | - Yao Wang
- Department of General Surgery, The People's Hospital of Ma Anshan, Ma Anshan, Anhui, China
| | - Eryun Gao
- Department of Oncology, The People's Hospital of Ma Anshan, Ma Anshan, Anhui, China,
| | - Fenglin Zhang
- Department of Oncology, The People's Hospital of Ma Anshan, Ma Anshan, Anhui, China,
| |
Collapse
|
57
|
Phung CD, Nguyen HT, Tran TH, Choi HG, Yong CS, Kim JO. Rational combination immunotherapeutic approaches for effective cancer treatment. J Control Release 2018; 294:114-130. [PMID: 30553850 DOI: 10.1016/j.jconrel.2018.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022]
Abstract
Immunotherapy is an important mode of cancer treatment. Over the past decades, immunotherapy has improved the clinical outcome for cancer patients. However, in many cases, mutations in cancer cells, lack of selectivity, insufficiency of tumor-reactive T cells, and host immunosuppression limit the clinical benefit of immunotherapy. Combination approaches in immunotherapy may overcome these obstacles. Accumulating evidence demonstrates that combination immunotherapy is the future of cancer treatment. However, designing safe and rational combinations of immunotherapy with other treatment modalities is critical. This review will discuss the optimal immunotherapy-based combinations mainly with respect to the mechanisms of action of individual therapeutic agents that target multiple steps in evasion and progression of tumor.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
58
|
PD-L1 Expression is Increased in Metastasizing Squamous Cell Carcinomas and Their Metastases. Am J Dermatopathol 2018; 40:647-654. [PMID: 29742559 DOI: 10.1097/dad.0000000000001164] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Programmed cell death ligand 1 (PD-L1) expression by tumor cells plays an important role in the inhibition of T cell-mediated immune response in cancer. PD-L1 expression by tumor cells has been linked to poor prognosis in a wide variety of cancers. However, PD-L1 expression in cutaneous squamous cell carcinoma (cSCC) has been scarcely studied, and its role as a prognosis biomarker remains controversial. The association of PD-L1 expression and the metastatic risk in a series of cSCC was assessed. PD-L1 and CD8 immunostainings of full excision sections of 99 primary tumors and 24 lymphatic metastases were semiquantitatively evaluated. Primary cSCCs were grouped according to the development of lymphatic metastatic spread [metastasizing squamous cell carcinoma (MSCC)] (n = 48) or the absence of progression [nonmetastasizing squamous cell carcinoma (NMSCC)] (n = 51). PD-L1-positive expression (cut off ≥1%) was found in 26% NMSCCs and in 50% MSCCs (P = 0.02). PD-L1 association with an increased metastatic risk was confirmed in the multivariate analysis (P < 0.05), along with the following features: recurrence, poor differentiation, and perineural invasion. Ninety percent of the metastases of PD-L1-positive tumors were also positive for PD-L1, displaying a trend toward a higher PD-L1 expression when compared with their primary tumors (P = 0.058). No significant differences in the peritumoral inflammatory infiltrate or in the expression of CD8 were found between metastasizing and nonmetastasizing primary tumors. Our results suggest that PD-L1 may play a relevant role in metastatic spread and may be a candidate prognostic biomarker in cSCC.
Collapse
|
59
|
Ma J, Li J, Qian M, Han W, Tian M, Li Z, Wang Z, He S, Wu K. PD-L1 expression and the prognostic significance in gastric cancer: a retrospective comparison of three PD-L1 antibody clones (SP142, 28-8 and E1L3N). Diagn Pathol 2018; 13:91. [PMID: 30463584 PMCID: PMC6249875 DOI: 10.1186/s13000-018-0766-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022] Open
Abstract
Background Immunohistochemistry (IHC) for programmed cell death ligand 1 (PD-L1) displays staining diversity. We compared IHC staining of PD-L1 in gastric cancer (GC) by using three commercially available antibody clones, and analyzed the correlation with the prognosis. Methods IHC using PD-L1 antibodies (clones SP142, 28–8 and E1L3N) in 315 formalin-fixed paraffin-embedded samples was qualitatively compared at the 1, 5 and 10% cut-off by two pathologists on total, tumor and immune/stromal cells. We used computer – assisted scoring to quantitatively analyze and compare the “H-score” of PD-L1 expression in 66 samples on total cells. The antibody clone SP142 was selected to investigate the infiltration of PD-L1+CD8+ T cells using automated quantitative immunofluorescence analyses (n = 50) and the prognostic significance. The prognoses were assessed by log-rank test. Results PD-L1 clones SP142 and 28–8 displayed great concordance by qualitative (κ = 0.816, 0.810 for total cells and tumor cells at the 5% cut-off) and quantitative analyses (R2 = 0.7991, 0.8187 for positive percentage and “H-score”). PD-L1 clone SP142 showed the highest positivity in immune/stromal cells staining (18.41%) compared to 28–8 (7.62%), while clone E1L3N showed poor staining in both tumor and immune/stromal cells. Clone SP142, but not 28–8 and E1L3N, predicted a worse prognosis at the 5% cut-off (p = 0.0243). Both the clone SP142 and 28–8 had high inter-pathologist correlation for tumor staining (R2 = 0.9805 and R2 = 0.9853), but a moderate correlation for stromal/immune cell staining (R2 = 0.5653 and R2 = 0.5745). Furthermore, a higher density of PD-L1+CD8+ T cells was correlated with a shorter survival time (R2 = 0.0909, p = 0.0352). Conclusions PD-L1 antibody clone SP142 was superior in cell staining, particularly in immune/stromal cell and prognosis. These findings are important for selection of PD-L1 antibody clones in the future diagnostic test. Electronic supplementary material The online version of this article (10.1186/s13000-018-0766-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing Ma
- Department of Gastroenterology, Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710032, Shaanxi Province, China.,State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jianhui Li
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Meirui Qian
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Weili Han
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Miaomiao Tian
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zengshan Li
- The Pathology Department, The Fourth Military Medical University, Xi'an, China
| | - Zhe Wang
- The Pathology Department, The Fourth Military Medical University, Xi'an, China
| | - Shuixiang He
- Department of Gastroenterology, Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710032, Shaanxi Province, China.
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
60
|
Duan J, Liu X, Chen H, Sun Y, Liu Y, Bai H, Wang J. Impact of PD-L1, transforming growth factor-β expression and tumor-infiltrating CD8 + T cells on clinical outcome of patients with advanced thymic epithelial tumors. Thorac Cancer 2018; 9:1341-1353. [PMID: 30168897 PMCID: PMC6209778 DOI: 10.1111/1759-7714.12826] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/04/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Advanced thymic epithelial tumors (TETs) are indolent and poorly responsive to chemotherapy. PD-1/PD-L1 inhibitors have shown remarkable clinical benefit in several cancers; however, many immunomodulatory molecules have been identified that affect the immune response. This study examined the progonostic roles of PD-L1, transforming growth factor-β (TGF-β), and CD8+ tumor-infiltrating lymphocytes (CD8+ TILs) in patients with TETs. METHODS Retrospective analysis was performed on the data of 20 patients with stage IV thymic carcinoma and 13 with stage III/IV invasive thymoma. Tissue biopsies were obtained before first-line chemotherapy was administered. Protein levels were assessed by immunohistochemistry. Objective response rate, overall survival (OS), and progression-free survival (PFS) were analyzed. RESULTS Patients with advanced thymic carcinoma exhibited higher levels of PD-L1 and TGF-β than patients with advanced invasive thymic carcinoma (PD-L1: 65.0% vs. 46.2%, P = 0.472; TGF-β: 65.0% vs. 15.4%, P = 0.011). Five advanced thymic carcinoma patients with low levels of PD-L1 and TGF-β exhibited high levels of CD8 staining. The median OS was 29.5 months patients with high TGF-β expression versus 62.9 in patients with low TGF-β (P = 0.052). In patients with advanced thymic carcinoma, the median PFS in the high PD-L1 expression group was 13.3 months versus 23.5 (P = 0.043) in the low PD-L1, and the median OS was 50.7 months in the high CD8 expression versus 15.1 in the CD8 low group (P = 0.154). CONCLUSIONS Our results showed the prognostic roles of PD-L1, TGF-β, and CD8+ TILs in patients with advanced TETs, and the potential for development of anti-PD-1/PD-L1 therapies.
Collapse
Affiliation(s)
- Jianchun Duan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xidong Liu
- Department of Interventional Oncology, Dong Ying People's Hospital, Dongying, China
| | - Han Chen
- Department of Thoracic Cancer, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yu Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yiqiang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hua Bai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
61
|
Guo J, Qu H, Shan T, Chen Y, Chen Y, Xia J. Tristetraprolin Overexpression in Gastric Cancer Cells Suppresses PD-L1 Expression and Inhibits Tumor Progression by Enhancing Antitumor Immunity. Mol Cells 2018; 41:653-664. [PMID: 29936792 PMCID: PMC6078856 DOI: 10.14348/molcells.2018.0040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/06/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022] Open
Abstract
The RNA-binding protein tristetraprolin (TTP) binds to adenosine-uridine AU-rich elements in the 3'-untranslated region of messenger RNAs and facilitates rapid degradation of the target mRNAs. Therefore, it regulates the expression of multiple cancer and immunity-associated transcripts. Furthermore, a lack of TTP in cancer cells influences cancer progression and predicts poor survival. Although the functions of TTP on cancer cells have previously been researched, the mechanism of TTP on the interaction between cancer cells with their microenvironment remains undiscovered. In this study, we admed to determine the role of cancer cell TTP during the interaction between tumor and immune cells, specifically regulatory T cells (Tregs). We evaluate the capability of TTP to modulate the antitumor immunity of GC and explored the underlying mechanism. The overexpression of TTP in GC cells dramatically increased peripheral blood mononuclear lymphocyte (PBML) -mediated cytotoxicity against GC cells. Increased cytotoxicity against TTP-overexpressed GC cells by PBMLs was determined by Treg development and infiltration. Surprisingly, we found the stabilization of programmed death-ligand 1 (PD-L1) mRNA was declining while TTP was elevated. The PD-L1 protein level was reduced in TTP-abundant GC cells. PD-L1 gas been found to play a pivotal role in Treg development and functional maintenance in immune system. Taken together, our results suggest the overexpression of TTP in GC cells not only affects cell survival and apoptosis but also increases PBMLs -mediated cytotoxicity against GC cells to decelerate tumor progression. Moreover, we identified PD-L1 as a critical TTP-regulated factor that contributes to inhibiting antitumor immunity.
Collapse
Affiliation(s)
- Jian Guo
- Department of General Surgery and Center of Translational Medicine, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University,
China
| | - Huiheng Qu
- Department of General Surgery and Center of Translational Medicine, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University,
China
| | - Ting Shan
- Department of General Surgery and Center of Translational Medicine, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University,
China
| | - Yigang Chen
- Department of General Surgery and Center of Translational Medicine, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University,
China
| | - Ye Chen
- Department of General Surgery and Center of Translational Medicine, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University,
China
| | - Jiazeng Xia
- Department of General Surgery and Center of Translational Medicine, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University,
China
| |
Collapse
|
62
|
Lin EM, Gong J, Klempner SJ, Chao J. Advances in immuno-oncology biomarkers for gastroesophageal cancer: Programmed death ligand 1, microsatellite instability, and beyond. World J Gastroenterol 2018; 24:2686-2697. [PMID: 29991874 PMCID: PMC6034145 DOI: 10.3748/wjg.v24.i25.2686] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/23/2018] [Accepted: 06/16/2018] [Indexed: 02/06/2023] Open
Abstract
Blockade of the programmed death ligand 1 (PD-L1) and programmed cell death 1 (PD-1) receptor axis represents an effective form of cancer immunotherapy. Preclinical evidence initially suggested that gastric and gastroesophageal junction (GEJ) cancers are potentially immunotherapy-sensitive tumors. Early phase clinical trials have demonstrated promising antitumor activity with PD-1/PD-L1 blockade in advanced or metastatic gastric/GEJ cancer. Microsatellite instability (MSI) and PD-L1 expression have been shown to predict higher response to PD-1 inhibitors as highlighted by the recent approvals of pembrolizumab in treatment-refractory solid tumors with MSI status and the third-line or greater treatment of PD-L1 positive advanced gastric/GEJ cancers. However, predictive and prognostic biomarkers remain an ongoing need. In this review, we detail the preclinical evidence and early tissue biomarker analyses illustrating potential predictive biomarkers to PD-1/PD-L1 blockade in gastric/GEJ cancer. We also review the clinical development of PD-1/PD-L1 inhibitors in gastric/GEJ cancer and highlight several areas in need of future investigation in order to optimize the efficacy of PD-1/PD-L1 blockade in gastric/GEJ cancer.
Collapse
Affiliation(s)
- Emily M Lin
- Department of Internal Medicine, Harbor-UCLA Medical Center, Torrance, CA 90509, United States
| | - Jun Gong
- Department of Medical Oncology and Developmental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, United States
| | - Samuel J Klempner
- The Angeles Clinic and Research Institute, Los Angeles, CA 90404, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Joseph Chao
- Department of Medical Oncology and Developmental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, United States
| |
Collapse
|
63
|
Wang X, Yang YE, Jia L, Chen Q. Gastroenterological Cancer and Immunotherapy. Can J Gastroenterol Hepatol 2018; 2018:4697670. [PMID: 30073155 PMCID: PMC6057351 DOI: 10.1155/2018/4697670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Xiaoping Wang
- Department of Pathology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yixin Eric Yang
- School of Natural Sciences, College of Natural, Applied and Health Sciences, Kean University, Union, NJ, USA
| | - Lintao Jia
- Department of Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, KS, USA
| |
Collapse
|
64
|
Yang Y, Alderman C, Sehlaoui A, Xiao Y, Wang W. MicroRNAs as Immunotherapy Targets for Treating Gastroenterological Cancers. Can J Gastroenterol Hepatol 2018; 2018:9740357. [PMID: 30046565 PMCID: PMC6038585 DOI: 10.1155/2018/9740357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 05/02/2018] [Indexed: 01/17/2023] Open
Abstract
Gastroenterological cancers are the most common cancers categorized by systems and are estimated to comprise 18.4% of all cancers in the United States in 2017. Gastroenterological cancers are estimated to contribute 26.2% of cancer-related death in 2017. Gastroenterological cancers are characterized by late diagnosis, metastasis, high recurrence, and being refractory to current therapies. Since the current targeted therapies provide limited benefit to the overall response and survival, there is an urgent need for developing novel therapeutic strategy to improve the outcome of gastroenterological cancers. Immunotherapy has been developed and underwent clinical trials, but displayed limited therapeutic benefit. Since aberrant expressions of miRNAs are found in gastroenterological cancers and miRNAs have been shown to regulate antitumor immunity, the combination therapy combining the traditional antibody-based immunotherapy and novel miRNA-based immunotherapy is promising for achieving clinical success. This review summarizes the current knowledge about the miRNAs and long noncoding RNAs that exhibit immunoregulatory roles in gastroenterological cancers and precancerous diseases of digestive system, as well as the miRNA-based clinical trials for gastroenterological cancers. This review also analyzes the ongoing challenge of identifying appropriate therapy candidates for complex and dynamic tumor microenvironment, ensuring efficient and targeted delivery to specific cancer tissues, and developing strategy for avoiding off-target effect.
Collapse
Affiliation(s)
- Yixin Yang
- College of Natural, Applied and Health Sciences, Kean University, 100 Morris Avenue, Union, NJ 07083, USA
| | - Christopher Alderman
- School of Medicine, University of Colorado, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Ayoub Sehlaoui
- Department of Biological Sciences, Emporia State University, 1 Kellogg Circle, Emporia, KS 66801, USA
| | - Yuan Xiao
- Department of Biological Sciences, Emporia State University, 1 Kellogg Circle, Emporia, KS 66801, USA
| | - Wei Wang
- Department of Thoracic Surgery III, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning 110042, China
| |
Collapse
|
65
|
Immunotherapy in Advanced Gastric Cancer: An Overview of the Emerging Strategies. Can J Gastroenterol Hepatol 2018; 2018:2732408. [PMID: 30027089 PMCID: PMC6031212 DOI: 10.1155/2018/2732408] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/11/2018] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) remains a public health problem, being the fifth most common cancer worldwide. In the western countries, the majority of patients present with advanced disease. Additionally, 65 to 75% of patients treated with curative intent will relapse and develop systemic disease. In metastatic disease, systemic treatment still represents the state of the art, with less than a year of median overall survival. The new molecular classification of GC was published in 2014, identifying four distinct major subtypes of gastric cancer, and has encouraged the investigation of new and more personalized treatment strategies. This paper will review the current evidence of immunotherapy in advanced gastric cancer.
Collapse
|
66
|
Wei M, Shen D, Mulmi Shrestha S, Liu J, Zhang J, Yin Y. The Progress of T Cell Immunity Related to Prognosis in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2018; 2018:3201940. [PMID: 29682534 PMCID: PMC5848132 DOI: 10.1155/2018/3201940] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023]
Abstract
Gastric cancer is the fifth most common malignancy all over the world, and the factors that can affect progress and prognosis of the gastric cancer patients are various, such as TNM stages, invasive depth, and lymph node metastasis ratio. T cell immunity is important component of human immunity system and immunity responding to tumor and dysfunction or imbalance of T cell immunity will lead to serious outcomes for body. T cell immunity includes many different types of cells, CD4+ T cell, CD8+ T cell, memory cell, and so on, and each of them has special function on antitumor response or tumor immune escape which is revealed in lung cancer, colorectal cancer, breast cancer, ovarian cancer, and so on. But its correlation with gastric cancer is not clear. Our review was preformed to explore the relationship between the progress and prognosis of gastric cancer (GC) and T cell immunity. According to recent researches, T cell immunity may have an important role in the progress and prognosis of GCs, but its function is affected by location, category, related molecule, and interaction between the cells, and some effects still are controversial. More researches are needed to clarify this correlation.
Collapse
Affiliation(s)
- Ming Wei
- Gastroenterology Department, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Duo Shen
- Gastroenterology Department, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Sachin Mulmi Shrestha
- Gastroenterology Department, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Juan Liu
- Gastroenterology Department, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Junyi Zhang
- Department of Critical Care Medicine, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Ying Yin
- Gastroenterology Department, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| |
Collapse
|
67
|
Ying L, Yan F, Meng Q, Yu L, Yuan X, Gantier MP, Williams BRG, Chan DW, Shi L, Tu Y, Ni P, Wang X, Chen W, Zang X, Xu D, Hu Y. PD-L1 expression is a prognostic factor in subgroups of gastric cancer patients stratified according to their levels of CD8 and FOXP3 immune markers. Oncoimmunology 2018; 7:e1433520. [PMID: 29872566 PMCID: PMC5980489 DOI: 10.1080/2162402x.2018.1433520] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/19/2022] Open
Abstract
Current studies aiming at identifying single immune markers with prognostic value have limitations in the context of complex antitumor immunity and cancer immune evasion. Here, we show how the integration of several immune markers influences the predictions of prognosis of gastric cancer (GC) patients. We analyzed Tissue Microarray (TMA) by multiplex immunohistochemistry and measured the expression of immune checkpoint molecule PD-L1 together with antitumor CD8 T cells and immune suppressive FOXP3 Treg cells in a cohort of GC patients. Unsupervised hierarchical clustering analysis of these markers was used to define correlations between CD8 T, FOXP3 Treg and PD-L1 cell densities. We found that FOXP3 and PD-L1 densities were elevated while CD8 T cells were decreased in tumor tissues compared to their adjacent normal tissues. However, patient stratification based on each one of these markers individually did not show significant prognostic value on patient survival. Conversely, combination of the ratios of CD8/FOXP3 and CD8/PD-L1 enabled the identification of patient subgroups with different survival outcomes. As such, high densities of PD-L1 in patients with high CD8/FOXP3 and low CD8/PD-L1 ratios correlated with increased survival. Collectively, this work demonstrates the need for the integration of several immune markers to obtain more meaningful survival prognosis and patient stratification. In addition, our work provides insights into the complex tumor immune evasion and immune regulation by the tumor-infiltrating effector and suppressor cells, informing on the best use of immunotherapy options for treating patients.
Collapse
Affiliation(s)
- Le Ying
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, P. R. China.,Department of Tea Science, Zhejiang University, Hangzhou, P. R. China
| | - Feng Yan
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, P. R. China
| | - Qiaohong Meng
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, P. R. China
| | - Liang Yu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, P. R. China
| | - Xiangliang Yuan
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Michael P Gantier
- Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Bryan R G Williams
- Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - David W Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Liyun Shi
- Department of Microbiology and Immunology, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yugang Tu
- Cell Signaling Technology, Inc., Asia Pacific
| | - Peihua Ni
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Xuefeng Wang
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Weisan Chen
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Victoria, Australia
| | - Xingxing Zang
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY; Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, P. R. China.,Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Yiqun Hu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
68
|
Böger C, Behrens HM, Mathiak M, Krüger S, Kalthoff H, Röcken C. PD-L1 is an independent prognostic predictor in gastric cancer of Western patients. Oncotarget 2018; 7:24269-83. [PMID: 27009855 PMCID: PMC5029700 DOI: 10.18632/oncotarget.8169] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 03/02/2016] [Indexed: 12/14/2022] Open
Abstract
Targeting the PD-1/PD-L1 immune checkpoint signaling is a novel promising treatment strategy in several tumor entities, and it is suggested that PD-L1/PD-1 expression is predictive for a PD-1/PD-L1 checkpoint inhibitor treatment response. We investigated the expression of PD-L1 and PD-1 by immunohistochemistry in a large and well characterized gastric cancer (GC) cohort of Caucasian patients, consisting of 465 GC samples and 15 corresponding liver metastases. Staining results were correlated with clinico-pathological characteristics and survival. PD-L1 expression was found in tumor cells of 140 GCs (30.1%) and 9 liver metastases (60%) respectively in immune cells of 411 GCs (88.4%) and 11 liver metastases (73.3%). PD-1 was expressed in tumor infiltrating lymphocytes in 250 GCs (53.8%) and in 11 liver metastases (73.3%). PD-L1 expression was significantly more prevalent in men, GCs of the proximal stomach, unclassified, papillary, Her2/neu-positive, Epstein-Barr-virus-positive, microsatellite instable, and PIK3CA-mutated GCs. A high PD-L1/PD-1 expression was associated with a significantly better patient outcome, and PD-L1 turned out to be an independent survival prognosticator. The correlation of PD-L1/PD-1 expression with distinct clinico-pathological patient characteristics may serve as a surrogate marker of PD-L1-positive GCs and may direct the use of immune checkpoint treatment strategies.
Collapse
Affiliation(s)
- Christine Böger
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | | | - Micaela Mathiak
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Sandra Krüger
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Holger Kalthoff
- Department of Experimental Cancer Research, Christian-Albrechts-University, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
69
|
Xiang X, Yu PC, Long D, Liao XL, Zhang S, You XM, Zhong JH, Li LQ. Prognostic value of PD -L1 expression in patients with primary solid tumors. Oncotarget 2018; 9:5058-5072. [PMID: 29435162 PMCID: PMC5797033 DOI: 10.18632/oncotarget.23580] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 12/13/2017] [Indexed: 01/05/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is thought to play a critical role in immune escape by cancer, but whether PD-L1 expression can influence prognosis of patients with solid tumors is controversial. Therefore, we meta-analyzed available data on whether PD-L1 expression correlates with overall survival (OS) in such patients. PubMed, EMBASE and other databases were systematically searched for cohort or case-control studies examining the possible correlation between PD-L1 expression and OS of patients with solid tumors. OS was compared between patients positive or negative for PD-L1 expression using scatter plots, and subgroup analyses were performed based on tumor type and patient characteristics. Data from 59 studies involving 20,004 patients with solid tumors were meta-analyzed. The median percentage of tumors positive for PD-L1 was 30.1%. OS was significantly lower in PD-L1-positive patients than in PD-L1-negative patients at 1 year (P = 0.039), 3 years (P < 0.001) and 5 years (P < 0.001). The risk ratios of OS (and associated 95% confidence intervals) were 2.02 (1.56-2.60) at 1 year, 1.57 (1.34-1.83) at 3 years and 1.43 (1.24-1.64) at 5 years. Similar results were obtained in subgroup analyses based on patient ethnicity or tumor type. The available evidence suggests that PD-L1 expression negatively affects the prognosis of patients with solid tumors. PD-L1 might serve as an efficient prognostic indicator in solid tumor and may represent the important new therapeutic target.
Collapse
Affiliation(s)
- Xiao Xiang
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Peng-Cheng Yu
- Department of Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Di Long
- Department of Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xiao-Li Liao
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Sen Zhang
- Department of Colorectal Anal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xue-Mei You
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jian-Hong Zhong
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| | - Le-Qun Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
70
|
Yang YQ, Dong WJ, Yin XF, Xu YN, Yang Y, Wang JJ, Yuan SJ, Xiao J, DeLong JH, Chu L, Xu HN, Zhou XM, Wang RW, Fang L, Liu XY, Zhang KJ. Interferon-related secretome from direct interaction between immune cells and tumor cells is required for upregulation of PD-L1 in tumor cells. Protein Cell 2018; 7:538-43. [PMID: 27295261 PMCID: PMC4930771 DOI: 10.1007/s13238-016-0281-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Yuan-Qin Yang
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wen-Jie Dong
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xiao-Fei Yin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan-Ni Xu
- College of Life Sciences, Northwest Agriculture and Forestry University, Yangling, 712100, China
| | - Yu Yang
- Central China Normal University, Wuhan, 430079, China
| | - Jiao-Jiao Wang
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Su-Jing Yuan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jing Xiao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jonathan Howard DeLong
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Liang Chu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hai-Neng Xu
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Xiu-Mei Zhou
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ru-Wei Wang
- Zhejiang Conba Pharmaceutical Co., Ltd, Hangzhou, 310018, China
| | - Ling Fang
- Zhejiang Conba Pharmaceutical Co., Ltd, Hangzhou, 310018, China
| | - Xin-Yuan Liu
- Xinyuan Institute of Medicine and Biotechnology, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Kang-Jian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- Sichuan Huiyang Life Science and Technology Corp., Chengdu, 610021, China.
| |
Collapse
|
71
|
Chakrabarti J, Holokai L, Syu L, Steele N, Chang J, Dlugosz A, Zavros Y. Mouse-Derived Gastric Organoid and Immune Cell Co-culture for the Study of the Tumor Microenvironment. Methods Mol Biol 2018; 1817:157-168. [PMID: 29959712 DOI: 10.1007/978-1-4939-8600-2_16] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The interaction between the receptor, programmed cell death protein 1 (PD-1) and ligand, programmed cell death 1 (PD-L1) is known to inhibit CD8+ cytotoxic T lymphocyte proliferation, survival, and effector function. The result of this interaction leads to evasion of immune surveillance by tumors and subsequently cancer cell proliferation. Immunotherapy via PD-L1 blockade is used for a variety of malignancies, yet the prognostic value of immune checkpoint inhibition for the treatment of gastric cancer remains controversial. Thus, preclinical models that would predict the efficacy of such therapy in a subgroup of gastric cancer patients would be an advancement in the personalized treatment of this disease. Three-dimensional organoid cultures have not only been used to investigate the mechanisms regulating development and disease, but have also been used for high-throughput drug screening for targeted personalized therapy. Here we present the methodology for the co-culture of mouse-derived gastric cancer organoids with autologous immune cells specifically for the study of PD-L1/PD-1 interactions within the tumor microenvironment in vitro.
Collapse
Affiliation(s)
- Jayati Chakrabarti
- Department of Pharmacology and System Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Loryn Holokai
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, OH, USA
| | - LiJyun Syu
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Nina Steele
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Julie Chang
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Andrzej Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yana Zavros
- Department of Pharmacology and System Physiology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
72
|
Tumor-associated macrophage infiltration is highly associated with PD-L1 expression in gastric adenocarcinoma. Gastric Cancer 2018; 21:31-40. [PMID: 28801853 DOI: 10.1007/s10120-017-0760-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/04/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Programmed death ligand 1 (PD-L1) is a key protein upregulated by tumor cells to suppress immune responses. Tumor-associated macrophages (TAMs) play a major role in this immunosuppression, but the relationship between PD-L1 expression and TAMs remains unclear in gastric adenocarcinoma (GAC). We simultaneously examined expression of PD-L1 and TAMs in GAC. METHODS We performed immunohistochemical staining for PD-L1, CD68 (pan-macrophage), and CD163 (M2-like macrophage) in 217 GAC samples using a tissue microarray. Expression of PD-L1 and CD68- and CD163-positive cells was evaluated using the Cytoplasmic V2.0 algorithm in Aperio ImageScope software, and logistic regression analysis was used to compare expression patterns between groups. RESULTS Thirty-one samples (14%) were positive for PD-L1 expression. The mean (± standard error) rates of infiltration were 6.83 ± 0.38% for CD68-positive cells and 6.16 ± 0.29% for CD163-positive cells. The mean rate of CD163-positive cell infiltration was significantly higher in diffuse GAC than in intestinal GAC (diffuse n = 111, 6.91%; intestinal n = 91, 5.26%; p = 0.006), but the mean rate of CD68-positive cell infiltration was similar between these types (p = 0.38). The mean infiltration rates of CD68- and CD163-positive cells in PD-L1-positive GAC were significantly higher than in PD-L1-negative GAC (CD68 p = 0.0002; CD163 p < 0.0001). In multivariate logistic regression analyses, CD163-positive cell infiltration was associated with PD-L1 expression (odds ratio 1.13; 95% confidence interval 1.02-1.25; p = 0.021). CONCLUSION M2-like macrophage infiltration is highly associated with PD-L1 expression in GAC cells, suggesting that macrophage infiltration can serve as a potential therapeutic target.
Collapse
|
73
|
Cho JH, Lee J. Anti-programmed death ligand 1 (PD-L1) inhibitors in metastatic gastric cancer. PRECISION AND FUTURE MEDICINE 2017. [DOI: 10.23838/pfm.2017.00177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
74
|
Kelly RJ. Immunotherapy for Esophageal and Gastric Cancer. Am Soc Clin Oncol Educ Book 2017; 37:292-300. [PMID: 28561677 DOI: 10.1200/edbk_175231] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PD-L1 upregulation occurs in approximately 40% of gastroesophageal cancers. However, unlike other solid tumors, there is minimal PD-L1 expressed on the cancer cells; rather, expression occurs predominantly on infiltrating myeloid cells. Preliminary clinical data involving single-agent PD-1/PD-L1 inhibitors in metastatic gastroesophageal cancer have reported response rates of 22%-27% for patients with PD-L1+ tumors and 10%-17% for unselected patients. The phase III ONO-4538-12 (ATTRACTION 2) trial has demonstrated an improved overall survival for nivolumab compared with placebo for patients with heavily pretreated gastric cancer. In the future, we will need better biomarkers to select those most likely to respond and/or identify patients who may need combination immunotherapeutics or alternate strategies. A number of subsets of gastric cancer with different immune signatures, most notably tumors positive for Epstein-Barr virus and microsatellite instability, have been identified, with approximately 50% and 94% PD-L1+ staining seen on tumor cells and immune cells in the EBV subtype and approximately 33% and 45% PD-L1+ staining seen on tumor cells and immune cells in MSI high tumors. Both subtypes demonstrate PD-L1+ immune cells with tumor-infiltrating patterns, unlike the more commonly seen PD-L1+ immune cells at the invasive margin. PD-L2 expression has been reported in 52% of esophageal adenocarcinomas but little is known about the expression of other immune checkpoints. Additional factors that suggest gastroesophageal cancers may respond to checkpoint inhibition include the high somatic mutation burden and the link with chronic inflammation. Here we provide a comprehensive review of the checkpoint inhibitor data published to date in advanced esophagogastric cancers and rationalize how the immune microenvironment in these diverse tumors can explain response or resistance to immunotherapeutics.
Collapse
Affiliation(s)
- Ronan J Kelly
- From The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
75
|
Abstract
Programmed death ligand 1 (PD-L1) is the principal ligand of programmed death 1 (PD-1), a coinhibitory receptor that can be constitutively expressed or induced in myeloid, lymphoid, normal epithelial cells and in cancer. Under physiological conditions, the PD-1/PD-L1 interaction is essential in the development of immune tolerance preventing excessive immune cell activity that can lead to tissue destruction and autoimmunity. PD-L1 expression is an immune evasion mechanism exploited by various malignancies and is generally associated with poorer prognosis. PD-L1 expression is also suggested as a predictive biomarker of response to anti-PD-1/PD-L1 therapies; however, contradictory evidence exists as to its role across histotypes. Over the years, anti-PD-1/PD-L1 agents have gained momentum as novel anticancer therapeutics, by inducing durable tumour regression in numerous malignancies including metastatic lung cancer, melanoma and many others. In this review, we discuss the immunobiology of PD-L1, with a particular focus on its clinical significance in malignancy.
Collapse
Affiliation(s)
- Anthousa Kythreotou
- Departmentof Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Abdul Siddique
- Departmentof Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Francesco A Mauri
- Departmentof Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Mark Bower
- National Centre for HIV Malignancy, Chelsea and Westminster Hospital, London, UK
| | - David J Pinato
- Departmentof Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| |
Collapse
|
76
|
Ju X, Shen R, Huang P, Zhai J, Qian X, Wang Q, Chen M. Predictive relevance of PD-L1 expression with pre-existing TILs in gastric cancer. Oncotarget 2017; 8:99372-99381. [PMID: 29245908 PMCID: PMC5725099 DOI: 10.18632/oncotarget.22079] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 10/11/2017] [Indexed: 12/29/2022] Open
Abstract
Expression of programmed cell death receptor ligand 1 (PD-L1) has been shown to be up-regulated in some gastric cancer patients and to correlate with the density of tumour infiltrating lymphocytes (TILs). However, conflicting results have been reported regarding TILs and the expression of PD-L1 as a prognostic marker for gastric cancer. We investigated the correlation of PD-L1 and TILs expression with clinicpathological characteristics in 105 well characterized gastric cancer patients. PD-L1 expression and CD3+ and CD8+ TILs were evaluated by fluorescent multiplex immunohistochemistry (mIHC) analysis. PD-L1 positive staining on tumour cells was observed in 35% cases and 48% cases showed PD-L1 expression on immune cells. Up-regulated PD-L1 expression on tumour cells and immune cells was associated with high density of pre-existing tumour infiltrating CD3+ and CD8+. In additional, more than 70% tumor infiltrating CD3+ cells were CD3+CD8+ cells. More than 60% PD-L1+ immune cells were PD-L1+CD3+CD8+ cells. PD-L1 expression in tumour cells was associated with poor prognosis and high density CD3+ and CD8+ TILs indicated improved overall survival in gastric cancer patients. Increased PD-L1 expression with low density CD3+ and CD8+ TILs had the shortest overall survival. In accordingly, PD-L1 absence with high density CD3+ and CD8+ TILs indicated the best prognosis. Combination of PD-L1 with pre-existing TILs may be more precise than PD-L1 alone for predicting survival in gastric cancer.
Collapse
Affiliation(s)
- Xiaoli Ju
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Rong Shen
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Pathology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Pan Huang
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jianhua Zhai
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaobin Qian
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qiang Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Miao Chen
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China.,Department of Pathology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
77
|
Ying L, Yan F, Meng Q, Yuan X, Yu L, Williams BRG, Chan DW, Shi L, Tu Y, Ni P, Wang X, Xu D, Hu Y. Understanding immune phenotypes in human gastric disease tissues by multiplexed immunohistochemistry. J Transl Med 2017; 15:206. [PMID: 29025424 PMCID: PMC5639762 DOI: 10.1186/s12967-017-1311-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022] Open
Abstract
Background Understanding immune phenotypes and human gastric disease in situ requires an approach that leverages multiplexed immunohistochemistry (mIHC) with multispectral imaging to facilitate precise image analyses. Methods We developed a novel 4-color mIHC assay based on tyramide signal amplification that allowed us to reliably interrogate immunologic checkpoints, including programmed death-ligand 1 (PD-L1), cytotoxic T cells (CD8+T) and regulatory T cells (Foxp3), in formalin-fixed, paraffin-embedded tissues of various human gastric diseases. By observing cell phenotypes within the disease tissue microenvironment, we were able to determine specific co-localized staining combinations and various measures of cell density. Results We found that PD-L1 was expressed in gastric ulcer and in tumor cells (TCs), as well as in tumor-infiltrating immune cells (TIICs), but not in normal gastric mucosa or other gastric intraepithelial neoplastic tissues. Furthermore, we found no significant reduction in CD8+T cells, whereas the ratio of CD8+T:Foxp3 cells and CD8+T:PD-L1 cells was suppressed in tumor tissues and elevated in adjacent normal tissues. An unsupervised hierarchical analysis also identified correlations between CD8+T and Foxp3+ tumor-infiltrating lymphocyte (TIL) densities and average PD-L1 levels. Three main groups were identified based on the results of CD8+T:PD-L1 ratios in gastric tumor tissues. Furthermore, integrating CD8+T:Foxp3 ratios, which increased the complexity for immune phenotype status, revealed 6–7 clusters that enabled the separation of gastric cancer patients at the same clinical stage into different risk-group subsets. Conclusions Characterizing immune phenotypes in human gastric disease tissues via multiplexed immunohistochemistry may help guide PD-L1 clinical therapy. Observing unique disease tissue microenvironments can improve our understanding of immune phenotypes and cell interactions within these microenvironments, providing the ability to predict safe responses to immunotherapies. Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1311-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Le Ying
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 227 Chongqing Road South, Shanghai, 200025, China.,Department of Tea Science, Zhejiang University, Hangzhou, China
| | - Feng Yan
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 227 Chongqing Road South, Shanghai, 200025, China.,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Qiaohong Meng
- Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Xiangliang Yuan
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 227 Chongqing Road South, Shanghai, 200025, China
| | - Liang Yu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated First People's Hospital, Shanghai, China
| | - Bryan R G Williams
- Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - David W Chan
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, P. R. China
| | - Liyun Shi
- Department of Microbiology and Immunology, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Yugang Tu
- Cell Signaling Technology, Inc., Asia Pacific, Danvers, USA
| | - Peihua Ni
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 227 Chongqing Road South, Shanghai, 200025, China
| | - Xuefeng Wang
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 227 Chongqing Road South, Shanghai, 200025, China. .,Institute of Ageing Research, Hangzhou Normal University School of Medicine, Hangzhou, China. .,Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia.
| | - Yiqun Hu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, 227 Chongqing Road South, Shanghai, 200025, China.
| |
Collapse
|
78
|
PD-L1 expression on immune cells is a favorable prognostic factor for vulvar squamous cell carcinoma patients. Oncotarget 2017; 8:89903-89912. [PMID: 29163797 PMCID: PMC5685718 DOI: 10.18632/oncotarget.20911] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/29/2017] [Indexed: 12/31/2022] Open
Abstract
Background Anti-immune programmed death-ligand 1 (PD-L1) pathway is used by the tumor to overcome immune system and serves as immunotherapy target in various malignancies. Aim To investigate the expression of PD-L1 in vulvar squamous cell carcinoma (vSCC) and to assess it's clinicopathological and prognostic significance. Methods Immunohistochemical PD-L1 expression was evaluated in 84 vSCCs with previously defined status of p16 and DNA-HPV, infiltration of immune cells: CD8+, CD4+, FOXP3+, CD56+, CD68+, and GZB+ cells. PD-L1 positivity was defined as ≥5% of PD-L1-positive cells. Survival analyses included the Kaplan–Meier method, log-rank test and Cox proportional hazards model. Results PD-L1 expression was detected on cancer and peritumoral immune cells. PD-L1-positivity of cancer nests (27/84, 32.1%) was correlated with higher infiltration of CD4+ (p=0.037), CD8+ (p=0.02), FOXP3+ (p=0.007), CD68+ (p=0.021) cells, while PD-L1 positivity of peritumoral immune cells (51/84, 60.7%) was correlated with higher infiltration of intraepithelial FOXP3+ cells only (p=0.037). PD-L1-positivity of cancer cells but not immune cells, was more frequently observed in p16-negative tumors (p=0.004). High-risk HPV-status did not correlate with the PD-L1 status of cancer and immune cells (p=1.000) and (p=1.000) respectively). Median follow up was 89.20 months (range 1.7-189.5). PD-L1 positivity of peritumoral immune cells was found to be an independent favorable prognostic factor for OS. Conclusion: This study highlights the importance of comprehensive PD-L1 assessment in both cancer and immune cells. PD-L1 expression on peritumoral immune cells seems to be an additional prognostic factor in vSCC patients and may influence the results by anti-PD-L1 treatment.
Collapse
|
79
|
Wu Y, Cao D, Qu L, Cao X, Jia Z, Zhao T, Wang Q, Jiang J. PD-1 and PD-L1 co-expression predicts favorable prognosis in gastric cancer. Oncotarget 2017; 8:64066-64082. [PMID: 28969052 PMCID: PMC5609984 DOI: 10.18632/oncotarget.19318] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 06/16/2017] [Indexed: 12/29/2022] Open
Abstract
While the prognosis of gastric cancer (GC) remains poor, PD-1 and PD-L1/L2 are promising prognostic biomarkers. We evaluated PD-1 and PD-L1/L2 expression in tumor cells (TCs) and tumor-infiltrating immune cells (TIICs). We determined the Helicobacter pylori (Hp) and Epstein-Barr virus (EBV) infection status in a GC cohort (n=340), then analyzed the relationship between the expression of PD-1, PD-L1/L2 and GC prognosis. We found that PD-1, PD-L1, and PD-L2 mRNA levels were up-regulated in GC tissues, and were positively correlated with one another (P=0.043, P=0.008 and P=0.035). PD-1 protein expression in TIICs was observed in 22.6% of GC patients. The PD-L1 and PD-L2 positivity rates were 40.3% and 53.8% in TCs, respectively, and 60.0% and 60.9% in TIICs, respectively. PD-L1 was up-regulated in EBV-infected GC patients in both TCs (P=0.009) and TIICs (P=0.003). Hp status was not associated with PD-1 or PD-L1/PD-L2 expression. In TIICs, PD-L1 expression was independently associated with better GC prognosis (HR=0.72, 95%CI: 0.53-0.99). Co-expression of PD-1 and PD-L1, but not PD-L2, was a favorable prognostic marker that indicated a dose effect on the mortality risk of GC patients (P-value for trend=0.005). Comprehensive evaluation of PD-1 and PD-L1 in TCs and TIICs could help predict the prognosis of gastric cancers, as well as reveal patients who might benefit from targeted treatment.
Collapse
Affiliation(s)
- Yanhua Wu
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Donghui Cao
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Limei Qu
- Department of Pathology, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zhifang Jia
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Tiancheng Zhao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Quan Wang
- Department of Gastric and Colorectal Surgery, First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jing Jiang
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
80
|
García-Pedrero JM, Martínez-Camblor P, Diaz-Coto S, Munguia-Calzada P, Vallina-Alvarez A, Vazquez-Lopez F, Rodrigo JP, Santos-Juanes J. Tumor programmed cell death ligand 1 expression correlates with nodal metastasis in patients with cutaneous squamous cell carcinoma of the head and neck. J Am Acad Dermatol 2017; 77:527-533. [DOI: 10.1016/j.jaad.2017.05.047] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 05/24/2017] [Accepted: 05/28/2017] [Indexed: 02/06/2023]
|
81
|
Cavalcanti E, Armentano R, Valentini AM, Chieppa M, Caruso ML. Role of PD-L1 expression as a biomarker for GEP neuroendocrine neoplasm grading. Cell Death Dis 2017; 8:e3004. [PMID: 28837143 PMCID: PMC5596583 DOI: 10.1038/cddis.2017.401] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/19/2017] [Accepted: 07/06/2017] [Indexed: 02/06/2023]
Abstract
Neuroendocrine neoplasms (NENs) are rare, heterogeneous and ubiquitous tumors commonly localized in the gastrointestinal tract, lung, and pancreas. The clinical behavior of NEN is highly unpredictable; in fact, low-grade cases can unexpectedly be associated with metastases. Currently, the 2010 WHO NEN classification employs histological differentiation and the proliferation index for grading tumors but fails to provide reliable prognostic and therapeutic indications. Therefore, there is an urgent need for a better characterization of G2/G3 NENs. Similar to several other tumors, NENs possess immune-escape mechanisms, but very little has yet been done to characterize this crucial aspect. There are no available data describing PD-L1 expression in these tumors. Here we provide, for the first time, evidence of PD-L1 tissue expression in gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). PD-L1 expression was significantly associated with a high-grade WHO classification (G3) (P<0.001) but not with gender, primary site, or lymph node status. The PD-L1 positivity rate and signal intensity are directly correlated (P<0.001) with a grade increase from G1 to G3. In particular in G3 cases, we observed a dichotomy between the morphology (WD- and PD-NENs) and Ki67. Moreover, our study demonstrated a significant association with the grade and PD-L1 expression levels in immune-infiltrating cells (P<0.001). In particular, G3 tumors are characterized by strong PD-L1 expression in both the tumor and infiltrating immune cells (P<0.001), reflecting an unfavorable environment for T-cell-mediated tumor aggression. These findings suggest that NENs might acquire resistance to immune surveillance by upregulating PD-L1 and inhibiting peritumoral and intratumoral infiltrating lymphocytes. Here we demonstrate that PD-L1 is currently the best-known biomarker for G3 NENs, becoming the new gold standard for G3 NEN discrimination. Furthermore, pharmacological approaches using anti-PD-1 antibodies may become the logical choice for the treatment of G3 cases with a poor prognosis.
Collapse
Affiliation(s)
- Elisabetta Cavalcanti
- Department of Pathology, National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Raffaele Armentano
- Department of Pathology, National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Anna Maria Valentini
- Department of Pathology, National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Marcello Chieppa
- Department of Pathology, National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy
| | - Maria Lucia Caruso
- Department of Pathology, National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Bari 70013, Italy
| |
Collapse
|
82
|
Cho J, Lee J, Bang H, Kim ST, Park SH, An JY, Choi MG, Lee JH, Sohn TS, Bae JM, Kang WK, Kim S, Kim KM. Programmed cell death-ligand 1 expression predicts survival in patients with gastric carcinoma with microsatellite instability. Oncotarget 2017; 8:13320-13328. [PMID: 28076847 PMCID: PMC5355099 DOI: 10.18632/oncotarget.14519] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is expressed in a subgroup of gastric cancers that may benefit from immunotherapy. Microsatellite instability-high (MSI-H) is a potential predictive factor for response to immunotherapy targeting the PD-1 or its ligand PD-L1. The relationship between PD-L1 expression and MSI-H status remains poorly understood. In this study, we investigated PD-L1 expression in patients with MSI-H gastric cancer. We analyzed PD-L1 expression in 78 MSI-H gastric cancer tissue samples using immunohistochemistry. PD-L1 expression was classified as expression on tumor cells or on immune cells. We observed PD-L1 expression in 48 gastric cancer samples (61.5%), consisting of 7 (9.0%) cases with tumor PD-L1 expression and 47 (60.3%) cases with immune cell PD-L1 expression. Immune cell PD-L1 expression was frequently associated with intestinal type cancer by the Lauren classification (p = 0.015), with a lower risk of lymph node metastasis (p = 0.027) and lower tumor stages (p = 0.029) compared to MSI-H gastric cancers without PD-L1 expression. Moreover, immune cell PD-L1 expression was an independent favorable prognostic factor for overall survival (versus PD-L1 negative; hazard ratio, 3.451; 95% confidence interval, 1.172–12.745; p = 0.025). In MSI-H gastric cancer, PD-L1 expression was observed to be independently associated with a longer survival.
Collapse
Affiliation(s)
- Junhun Cho
- Department of Pathology, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Heejin Bang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Tae Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se Hoon Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Yeong An
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Min Gew Choi
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Ho Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Sung Sohn
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Moon Bae
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Ki Kang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
83
|
Gu L, Chen M, Guo D, Zhu H, Zhang W, Pan J, Zhong X, Li X, Qian H, Wang X. PD-L1 and gastric cancer prognosis: A systematic review and meta-analysis. PLoS One 2017; 12:e0182692. [PMID: 28796808 PMCID: PMC5552131 DOI: 10.1371/journal.pone.0182692] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022] Open
Abstract
The expression of Programmed cell Death Ligand 1 (PD-L1) is observed in many malignant tumors and is associated with poor prognosis including Gastric Cancer (GC). The relationship between PD-L1 expression and prognosis, however, is controversial in GC. This paper purports to use a meta-analysis to investigate the relationship between PD-L1 expression and prognosis in GC. For this study, the following databases were searched for articles published from June 2003 until February 2017: PubMed, EBSCO, Web of Science and Cochrane Library. The baseline information extracted were: authors, year of publication, country where the study was performed, study design, sample size, follow-up time, baseline characteristics of the study population, pathologic data, overall survival (OS). A total of 15 eligible studies covering 3291 patients were selected for a meta-analysis based on specified inclusion and exclusion criteria. The analysis showed that the expression level of PD-L1 was associated with the overall survival in GC (Hazard Ratio, HR = 1.46, 95%CI = 1.08-1.98, P = 0.01, random-effect). In addition to the above, subgroup analysis showed that GC patients with deeper tumor infiltration, positive lymph-node metastasis, positive venous invasion, Epstein-Barr virus infection positive (EBV+), Microsatellite Instability (MSI) are more likely to expression PD-L1. The results of this meta-analysis suggest that GC patients, specifically EBV+ and MSI, may be prime candidates for PD-1 directed therapy. These findings support anti-PD-L1/PD-1 antibodies as a kind of immunotherapy which is promising for GC.
Collapse
Affiliation(s)
- Lihu Gu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Manman Chen
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dongyu Guo
- Department of Ophthalmology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hepan Zhu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenchao Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junhai Pan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Zhong
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinlong Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haoran Qian
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xianfa Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
84
|
Luo X, Peng X, Hou J, Wu S, Shen J, Wang L. Folic acid-functionalized polyethylenimine superparamagnetic iron oxide nanoparticles as theranostic agents for magnetic resonance imaging and PD-L1 siRNA delivery for gastric cancer. Int J Nanomedicine 2017; 12:5331-5343. [PMID: 28794626 PMCID: PMC5536232 DOI: 10.2147/ijn.s137245] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Programmed death ligand-1 (PD-L1), which is highly expressed in gastric cancers, interacts with programmed death-1 (PD-1) on T cells and is involved in T-cell immune resistance. To increase the therapeutic safety and accuracy of PD-1/PD-L1 blockade, RNA interference through targeted gene delivery was performed in our study. We developed folic acid (FA)- and disulfide (SS)-polyethylene glycol (PEG)-conjugated polyethylenimine (PEI) complexed with superparamagnetic iron oxide Fe3O4 nanoparticles (SPIONs) as a siRNA-delivery system for PD-L1 knockdown. The characterization, binding ability, cytotoxicity, transfection efficiency, and cellular internalization of the polyplex were determined. At nitrogen:phosphate (N:P) ratios of 10 or above, the FA-PEG-SS-PEI-SPIONs bound to PD-L1 siRNA to form a polyplex with a diameter of approximately 120 nm. Cell-viability assays showed that the polyplex had minimal cytotoxicity at low N:P ratios. The FA-conjugated polyplex showed higher transfection efficiency and cellular internalization in the folate receptor-overexpressing gastric cancer cell line SGC-7901 than a non-FA-conjugated polyplex. Subsequently, we adopted the targeted FA-PEG-SS-PEI-SPION/siRNA polyplexes at an N:P ratio of 10 for function studies. Cellular magnetic resonance imaging (MRI) showed that the polyplex could also act as a T2-weighted contrast agent for cancer MRI. Furthermore, one of four PD-L1 siRNAs exhibited effective PD-L1 knockdown in PD-L1-overexpressing SGC-7901. To determine the effects of the functionalized polyplex on T-cell function, we established a coculture model of activated T cells and SGC-7901 cells and demonstrated changes in secreted cytokines. Our findings highlight the potential of this class of multifunctional theranostic nanoparticles for effective targeted PD-L1-knockdown therapy and MRI diagnosis in gastric cancers.
Collapse
Affiliation(s)
- Xin Luo
- Department of Gastroenterology
| | | | - Jingying Hou
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital
| | - Shuyun Wu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
| | - Jun Shen
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | | |
Collapse
|
85
|
Dai C, Wang M, Lu J, Dai Z, Lin S, Yang P, Tian T, Liu X, Min W, Dai Z. Prognostic and predictive values of PD-L1 expression in patients with digestive system cancer: a meta-analysis. Onco Targets Ther 2017; 10:3625-3634. [PMID: 28769571 PMCID: PMC5533487 DOI: 10.2147/ott.s138044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND PD-L1 has been reported to be expressed in diverse human malignancies. However, the prognostic value of PD-L1 in digestive system cancers remains inconclusive. Therefore, we conducted this meta-analysis to evaluate the prognostic impact of PD-L1 expression in digestive system cancers. MATERIALS AND METHODS We searched the PubMed, Embase, and the Chinese National Knowledge Infrastructure for publications concerning PD-L1 expression in digestive system cancers. Correlations of PD-L1 expression level with overall survival (OS), disease-free survival (DFS), and recurrence-free survival (RFS) were analyzed. RESULTS Finally, 32 studies with 7,308 patients were included. Our results show that PD-L1 expression was significantly associated with poorer OS (hazard ratio [HR] =1.44, 95% confidence interval [CI] =1.18-1.76, P<0.001), but not DFS (HR =0.91, 95% CI =0.61-1.37, P=0.657) or RFS (HR =1.27, 95% CI =0.75-2.14, P=0.368). Moreover, in the subgroup analysis, significant associations between PD-L1 expression and OS were found in Asians (HR =1.50, 95% CI =1.19-1.89, P=0.001), gastric cancer (HR =1.43, 95% CI =1.05-1.94, P=0.021), and pancreatic carcinoma (HR =2.64, 95% CI =1.78-3.93, P<0.001). CONCLUSION These results suggest that the expression of PD-L1 is associated with worse OS in digestive system cancers, especially in gastric cancer and pancreatic cancer. In addition, PD-L1 may act as a new parameter for predicting poor prognosis and a promising target for anticancer therapy in digestive system cancers.
Collapse
Affiliation(s)
- Cong Dai
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Jun Lu
- Clinical Research Center, The First Affiliated Hospital of Xi’an Jiaotong University
| | - Zhiming Dai
- Department of Anesthesia, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Pengtao Yang
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Tian Tian
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Xinghan Liu
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Weili Min
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi’an Jiaotong University
| |
Collapse
|
86
|
Programmed Death-Ligand 1 Expression Is Common in Gastric Cancer Associated With Epstein-Barr Virus or Microsatellite Instability. Am J Surg Pathol 2017; 40:1496-1506. [PMID: 27465786 DOI: 10.1097/pas.0000000000000698] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Blockade of the programmed death 1 (PD-1) pathway has emerged as a novel therapy for cancer. Therefore, development of biomarkers for response prediction, such as PD-ligand 1 (PD-L1) expression by immunohistochemistry, may help to stratify patients. Solid tumors with CD8 T-cell rich tumor microenvironment have been implicated to be associated with increased PD-L1 expression. We hypothesized that gastric cancers associated with Epstein-Barr virus infection (EBV+) or microsatellite instability (MSI), both of which are known to harbor such tumor microenvironment, are associated with increased PD-L1 expression. Forty-four resected gastric cancers including 7 EBV+, 16 MSI, and 21 microsatellite stable cancers without EBV (EBV-/MSS) were studied for PD-L1 expression and T-cell subpopulations by immunohistochemistry. Positive PD-L1 expression (PD-L1+), defined as membranous staining in either tumor cells or tumor immune infiltrates, was seen in 32 (72%) gastric cancers. EBV+ or MSI cancers showed significantly higher rates of PD-L1+ compared with EBV-/MSS cancers (7/7, 100%; 14/16, 87%; 11/21, 52%; P=0.013). PD-L1+/EBV+ and PD-L1+/MSI cancers had significantly more CD8 T cells at tumor invasive front than PD-L1+/EBV-/MSS cancers (P<0.001). PD-L1+ was not associated with the depth of invasion or nodal metastasis (P=0.534, 0.288). Multivariate analysis showed PD-L1+ was not an independent predictor of disease-free survival while MSI was (P=0.548, 0.043). In summary, EBV+ or MSI gastric cancers are more likely to express PD-L1 and have increased CD8 T cells at tumor invasive front than EBV-/MSS cancers. Our results suggest EBV infection and MSI should be investigated for predicting response to PD-1 blockade.
Collapse
|
87
|
Que Y, Xiao W, Guan YX, Liang Y, Yan SM, Chen HY, Li QQ, Xu BS, Zhou ZW, Zhang X. PD-L1 Expression Is Associated with FOXP3+ Regulatory T-Cell Infiltration of Soft Tissue Sarcoma and Poor Patient Prognosis. J Cancer 2017; 8:2018-2025. [PMID: 28819402 PMCID: PMC5559963 DOI: 10.7150/jca.18683] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/01/2017] [Indexed: 02/07/2023] Open
Abstract
Background: Programmed death ligand-1(PD-L1) functions as a negative mediator of immune response through different pathways in anti-tumor immunity. Recent studies have reported that PD-L1 plays a pivotal role in the function of regulatory T-cells (Tregs). Although increases in FOXP3+ Tregs infiltration and PD-L1 expression have been revealed in several cancers, their correlation with soft tissue sarcoma remains unknown. Methods: We included 163 cases of soft tissue sarcoma who were diagnosed and underwent extensive and radical resection at the Sun Yat-sen University Cancer Center, Guangzhou, China, from 2000-2010. PD-L1 and FOXP3 expression was evaluated by immunohistochemistry. Correlation between their expressions and associations with clinicopathological features were studied. Results: Among 163 STS samples, 19 (11.7%) exhibited PD-L1 positivity, and 41 (25.2%) cases expressed high FOXP3+ Treg infiltration. Significant correlation between PD-L1 expression and FOXP3+Treg infiltration in STS was identified (r=0.450, p<0.001). In univariate analysis, PD-L1 expression was significantly associated with high tumor grade and the age of patients, while the presence of FOXP3+ in tumor infiltrating Tregs was significantly associated with the age of patients, high tumor stage, higher tumor grade and tumor depth. Multivariate analysis revealed PD-L1 and FOXP3 as independent prognostic indicators significantly associated with OS and DFS. Conclusions: Our study revealed that PD-L1 and FOXP3+Tregs may work synergistically in promoting immune evasion of the tumors in soft tissue sarcoma. A combined strategy to block PD-L1/PD-1 with simultaneous depletion of Tregs may show promise in enhancing the therapeutic efficacy of these patients.
Collapse
Affiliation(s)
- Yi Que
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Wei Xiao
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Yuan-Xiang Guan
- Department of Gastric and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Yao Liang
- Department of Gastric and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Shu-Mei Yan
- Department of Pathology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Huo-Ying Chen
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Qiao-Qiao Li
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Bu-Shu Xu
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Zhi-Wei Zhou
- Department of Gastric and Pancreatic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| | - Xing Zhang
- Department of Medical Melanoma and Sarcoma, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 East Dongfeng Road, Guangzhou 510060, China
| |
Collapse
|
88
|
The Human Stomach in Health and Disease: Infection Strategies by Helicobacter pylori. Curr Top Microbiol Immunol 2017; 400:1-26. [PMID: 28124147 DOI: 10.1007/978-3-319-50520-6_1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori is a bacterial pathogen which commonly colonizes the human gastric mucosa from early childhood and persists throughout life. In the vast majority of cases, the infection is asymptomatic. H. pylori is the leading cause of peptic ulcer disease and gastric cancer, however, and these outcomes occur in 10-15% of those infected. Gastric adenocarcinoma is the third most common cause of cancer-associated death, and peptic ulcer disease is a significant cause of morbidity. Disease risk is related to the interplay of numerous bacterial host and environmental factors, many of which influence chronic inflammation and damage to the gastric mucosa. This chapter summarizes what is known about health and disease in H. pylori infection, and highlights the need for additional research in this area.
Collapse
|
89
|
Fang W, Chen Y, Sheng J, Zhou T, Zhang Y, Zhan J, Liu L, Huang J, Peng P, Zhang L. Association between PD-L1 Expression on Tumour-Infiltrating Lymphocytes and Overall Survival in Patients with Gastric Cancer. J Cancer 2017; 8:1579-1585. [PMID: 28775777 PMCID: PMC5535713 DOI: 10.7150/jca.18729] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/31/2017] [Indexed: 12/14/2022] Open
Abstract
Purpose Targeting of the PD-1/PD-L1 signalling pathway is a promising treatment strategy in several cancers. The aim of this study was to assess the expression of PD-L1 on tumour cells and tumour-infiltrating lymphocytes (TILs) in gastric cancer (GC) and its prognostic impact. Materials and Methods A total of 240 patients who were diagnosed with GC at Sun Yat-sen University Cancer Centre (SYSUCC) from May 2008 to December 2013 were included in this study. PD-L1 expression was detected by immunohistochemistry (IHC) in all GC tumour specimens. The Cox proportional hazard regression model was used to assess the association between PD-L1 expression and overall survival (OS). Results The positive rates of PD-L1 expression on tumour cells and TILs were 74.8% and 65.8%, respectively. Patients with poor tumour differentiation had higher positive rates of PD-L1 expression on tumour cells (p=0.023). There was no significant association between PD-L1 expression on tumour cells and other clinicopathological data. In TILs, PD-L1 expression was significantly higher in patients who underwent surgery (p=0.031) and were in the late stage (p=0.021) than those without surgery and in the early stage. Patients with positive PD-L1 expression on TILs had a significantly shorter five-year OS than those with negative PD-L1 expression (14.2 vs 18.3; p=0.001); therefore, PD-L1 expression on TILs is an independent prognostic factor. However, PD-L1 expression on tumour cells is not associated with OS (p=0.945). Conclusion Our findings suggest that PD-L1 expression on TILs may be a predictive factor for immunotherapy of PD-1/PD-L1 pathway inhibitors.
Collapse
Affiliation(s)
- Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying Chen
- Department of Medical Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhu Hai, China
| | - Jin Sheng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ting Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yaxiong Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jianhua Zhan
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lin Liu
- Department of Medical Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhu Hai, China
| | - Jiaxing Huang
- Department of Medical Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhu Hai, China
| | - Peijian Peng
- Department of Medical Oncology, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhu Hai, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Guangzhou, China.,Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
90
|
Abstract
Gastrointestinal (GI) cancers such as gastric, esophageal, pancreas, hepatobiliary, colorectal and anal cancers are a major cause of cancer related mortality worldwide. Traditional treatment options such as chemotherapy, surgery, radiation therapy, monoclonal antibodies and anti-angiogenic agents have been the backbone of treatment of GI cancers in various stages. Current cancer research is moving forward to incorporate immunotherapies in the treatment of GI cancers either as single agent or in combination with current available treatment modalities. This review summarizes the existing and ongoing immunotherapies in the treatment of GI cancers.
Collapse
Affiliation(s)
- Patrick Grierson
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kian-Huat Lim
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Manik Amin
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
91
|
Abstract
BACKGROUND An increasing number of studies have examined the ability of programmed death-ligand 1 (PD-L1) to function as a marker for tumor prognosis. However, whether PD-L1 expression is a prognostic factor for the poor outcomes in many human cancers remains controversial. This study aims to investigate the prognostic role of PD-L1 expression through a meta-analysis update of 60 studies. METHODS The studies were identified by searching PubMed, Embase, Google Scholar, and Cochrane Library, and were assessed by further quality evaluation. The pooled hazard ratios (HRs) with 95% confidence intervals (CIs) for total and stratified analyses were calculated to investigate the association between the PD-L1 expression and the overall (OS) and disease-free (DFS) or progression-free survivals (PFS) of cancer patients. Heterogeneity and publication bias were also investigated. RESULTS The results indicated that PD-L1 overexpression can predict a poor OS (HR = 1.58, 95% CI = 1.38-1.81, P <.000) and DFS/PFS (HR = 1.72, 95% CI = 1.26-2.33, P = .001). Subgroup analyses showed that PD-L1 overexpression was significantly related to the poor OS in patients with breast (HR = 1.98, 95% CI = 1.15-3.41, P = .014), urothelial (HR = 2.24, 95% CI = 1.61-3.12, P <.000), renal (HR = 3.30, 95% CI = 2.23-4.86, P <.000), and gastric cancers (HR = 1.56, 95% CI = 1.02-2.37, P = .040). Furthermore, PD-L1 overexpresion was significantly associated with poor DFS/PFS in patients with hepatocellular carcinoma (HCC) (HR = 1.72, 95% CI = 1.21-2.46, P = .003), melanoma (HR = 3.39, 95% CI = 2.02-5.69, P <.000), and renal carcinoma, (HR = 5.04, 95% CI = 2.87-8.86, P <.000). The adverse prognostic impact of PD-L1 was observed in patients of different ethnicities. CONCLUSIONS The findings of this meta-analysis suggest the correlation of PD-L1 overexpression with worse OS in patients with solid tumors. However, the correlations differed according to tumor types.
Collapse
Affiliation(s)
| | - Fang Liu
- Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
92
|
Zhao T, Li C, Wu Y, Li B, Zhang B. Prognostic value of PD-L1 expression in tumor infiltrating immune cells in cancers: A meta-analysis. PLoS One 2017; 12:e0176822. [PMID: 28453554 PMCID: PMC5409185 DOI: 10.1371/journal.pone.0176822] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/18/2017] [Indexed: 01/28/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a promising target of cancer immune therapy. It not only expressed in tumor cells (TCs) but also up regulated in tumor infiltrating immune cells (TIICs). Although the previous meta-analysis have shown that PD-L1 expression in TCs was a valuable biomarker in predicting cancer prognosis, but few researches systematic evaluated the association between its expression in TIICs and survival of cancer patients. Thus, we performed this meta-analysis to evaluate the prognostic value of PD-L1 expression in TIICs in different types of cancers. Our results are valuable supplements when using PD-L1 expression to predict the survival of cancer patients and to select the beneficial patients from PD-L1 target therapy. PubMed, Embase, Web of Science and the Cochrane Central Search Library were used to perform our systematic literature search. Overall survival (OS) at 5th years and hazard ratios (HRs) were calculated using random effects models. Eighteen studies involving 3674 patients were included. The median positive rate of PD-L1 staining in TIICs was 36.37%. PD-L1 positive expression in TIICs related to a lower risk of death (HR = 0.784, 95%CI: 0.616–0.997, P = 0.047). Subgroup analyses found that PD-L1 positive expression in TIICs indicated a better prognosis especially in breast cancer patients (HR = 0.359, P = 0.041). When using whole tissue section slides, or using ‘any expression in TIICs’ as a cutoff value to assessing the results of IHC staining, PD-L1 expression in TIICs had a good prognostic value in cancer prognosis (HR = 0.587, P = 0.001 and HR = 0.549, P = 0.002). Our findings suggested that PD-L1 expression in TIICs was related to a better survival of cancer. The comprehensive evaluation of tumor cells and tumor infiltrating immune cells are required when evaluating the effect of PD-L1 expression on prognosis of cancer in future research.
Collapse
Affiliation(s)
- Tiancheng Zhao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yanhua Wu
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, Jilin, China
- * E-mail: (BZ); (BJL)
| | - Bin Zhang
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
- * E-mail: (BZ); (BJL)
| |
Collapse
|
93
|
Bilgin B, Sendur MAN, Bülent Akıncı M, Şener Dede D, Yalçın B. Targeting the PD-1 pathway: a new hope for gastrointestinal cancers. Curr Med Res Opin 2017; 33:749-759. [PMID: 28055269 DOI: 10.1080/03007995.2017.1279132] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND VEGF, HER2 and EGFR targeted agents are currently used in gastric, esophageal and colorectal cancers. However, treatment outcomes are still poor in most gastrointestinal (GI) cancers. Immune checkpoints are one of the most promising immunotherapy approaches. In this review article, we aim to discuss the efficacy and safety of anti-PD-1/PD-L1 therapies in GI cancers, including gastric, esophageal and colorectal cancer in published or reported recent studies. SCOPE A literature search was made from PubMed and ASCO Annual Meeting abstracts by using the following search keywords: "nivolumab", "pembrolizumab", "avelumab", "GI cancers" "anti-PD1 therapy" and "anti-PD-L1 therapy". The last search was on 2 November 2016. The most important limitation of our review is that most of the data on anti-PD-1/PD-L1 therapies in GI cancers relies on phase 1 and 2 trials. FINDINGS Currently, there are two anti-PD-1 (nivolumab and pembrolizumab) and one anti-PDL1 (atezolizumab) agents approved by FDA. After the treatment efficacy of immune checkpoint blockade was shown in melanoma, renal cell cancer and non-squamous lung cancer, trials which evaluate immune checkpoint blockade in GI cancers are ongoing. Early results of trials have been promising and encouraging for patients with advanced stage gastroesophageal cancer. According to early results of published trials, response to anti-PD1/PD-L1 agents appears to be associated with tumor PD-L1 levels. According to two recently published phase 2 trials, the clinical benefits of immune checkpoint blockade with both nivolumab and pembrolizumab were limited in patients with microsatellite instability (MSI) positive advanced colorectal cancer. However, several phase 2/3 trials are still ongoing. CONCLUSION Both pembrolizumab and nivolumab show promising efficacy with acceptable safety data in published trials in GI cancers, especially in refractory MSI positive metastatic colorectal cancer.
Collapse
Affiliation(s)
- Burak Bilgin
- a Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Mehmet A N Sendur
- a Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Muhammed Bülent Akıncı
- a Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Didem Şener Dede
- a Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| | - Bülent Yalçın
- a Yıldırım Beyazıt University , Faculty of Medicine, Department of Medical Oncology , Ankara , Turkey
| |
Collapse
|
94
|
Murphy AG, Kelly RJ. The Evolving Role of Checkpoint Inhibitors in the Management of Gastroesophageal Cancer. Hematol Oncol Clin North Am 2017; 31:485-498. [PMID: 28501089 DOI: 10.1016/j.hoc.2017.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The connection between inflammation and malignancy has long been recognized in gastric and esophageal cancers. Given the considerable success of immune checkpoint inhibitors in other tumor types, for example, lung cancer and melanoma, much attention is being paid to furthering their role in gastric and esophageal cancers. The Cancer Genome Atlas has provided further details of the molecular heterogeneity of these tumors, which may help predict responsiveness to immune checkpoint inhibitors. This article discusses the rationale for investigating these agents in gastroesophageal cancer and summarizes the relevant clinical trial data and ongoing studies.
Collapse
Affiliation(s)
- Adrian G Murphy
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, The Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Room G93, Baltimore, MD 21231, USA
| | - Ronan J Kelly
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, The Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Room G93, Baltimore, MD 21231, USA.
| |
Collapse
|
95
|
Fukuda K, Yoshida T, Shimazu K, Taguchi D, Inoue M, Nanjyo H, Shibata H. Effect of anti-PD-1 antibody, nivolumab on early gastric cancer. Int Cancer Conf J 2017; 6:98-103. [PMID: 31149480 DOI: 10.1007/s13691-017-0284-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/06/2017] [Indexed: 02/11/2025] Open
Abstract
Immunomodulation treatment using anti-programmed cell death protein 1 antibody is a very promising treatment for various types of advanced cancers, including melanoma, non-small cell lung cancer, and renal cell carcinoma. However, the therapeutic effects on early cancers are still unknown. We experienced 2 cases of early gastric cancers coexisting advanced melanomas. In both cases, early gastric cancers did not respond to nivolumab. Both early gastric cancers did not express programed death ligand 1. It was speculated that immunotolerance was not fully established responsible for nivolumab in early gastric cancer, and for this reason, nivolumab could not shrink these tumors. We experienced 2 cases of early gastric cancer, where PD-L1 was negative, not responding to nivolumab.
Collapse
|
96
|
Prognostic role of PD-L1 in malignant solid tumors: a meta-analysis. Int J Biol Markers 2017; 32:e68-e74. [PMID: 27470134 DOI: 10.5301/jbm.5000225] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2016] [Indexed: 12/20/2022]
Abstract
PURPOSE The aim of this study was to elucidate the rates and prognostic roles of programmed cell death ligand 1 (PD-L1) immunohistochemical (IHC) expression in various malignant tumors through a systematic review and meta-analysis. METHOD The current study included 16,176 patients from 97 eligible studies. We investigated PD-L1 expression and its correlation with survival rate in various malignant tumors. RESULTS The estimated rate of PD-L1 IHC expression was 0.449 (95% confidence interval [CI] 0.404-0.495). The highest and lowest PD-L1 expression levels were found in thyroid cancer (0.829, 95% CI 0.781-0.868) and small-cell neuroendocrine carcinoma (0.005, 95% CI 0.000-0.080), respectively. PD-L1 expression was significantly correlated with poorer overall survival and disease-free survival rates (hazard ratios 1.276, 95% CI 1.097-1.486 and 1.304, 95% CI 1.034-1.644, respectively). However, PD-L1 IHC expression was significantly correlated with worse overall survival rates in patients with esophageal cancer and renal cell carcinoma and with worse disease-free survival rates in patients with colorectal cancer, hepatocellular carcinoma, and renal cell carcinoma. CONCLUSIONS Our results show that PD-L1 expression rates and the correlations with survival varied between tumor types. Detailed evaluation criteria for PD-L1 will have to be standardized before application to specific tumor types.
Collapse
|
97
|
Giampieri R, Maccaroni E, Mandolesi A, Del Prete M, Andrikou K, Faloppi L, Bittoni A, Bianconi M, Scarpelli M, Bracci R, Scartozzi M, Cascinu S. Mismatch repair deficiency may affect clinical outcome through immune response activation in metastatic gastric cancer patients receiving first-line chemotherapy. Gastric Cancer 2017; 20:156-163. [PMID: 26796888 DOI: 10.1007/s10120-016-0594-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/09/2016] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The microsatellite-instable gastric cancer subtype, because of its supposed high antigenic potential, is a promising candidate for immunotherapy. We analyzed if the presence of a defective mismatch repair (MMR) system is associated with other markers of immune response and their relationship with outcome in advanced gastric cancer patients. METHODS We analyzed the relationship between clinical outcome and MMR status, the presence of tumor-infiltrating lymphocytes (TIL), lymphocytosis, and neutrophil-to-lymphocyte ratio (NLR) in metastatic gastric cancer patients treated with a chemotherapy doublet in the first-line setting. Other stratification factors were sex, age, Eastern Cooperative Oncology Group performance status, adjuvant/neoadjuvant chemotherapy, metastatic sites, and histotype. RESULTS One hundred three patients were eligible for analysis. Defective MMR was found in 15 patients (14 %), TILs were found in 18 patients (17 %), lymphocytosis was found in 24 patients (23 %), and high NLR was found in 75 patients (72 %). Significant correlations were found between defective MMR and TIL positivity (p = 0.0004), between defective MMR and lymphocytosis (p = 0.0062), between defective MMR and low NLR (p = 0.000069), and between TIL positivity and lymphocytosis (p = 0.000147). All factors had a statistically significant impact on overall survival, although on multivariate analysis only defective MMR (p = 0.0001) and TIL positivity (p = 0.0192) maintained their independent prognostic role. Similar results were observed for progression-free survival, with defective MMR (p = 0.0001) and TIL positivity (p = 0.0195) maintaining their prognostic role on multivariate analysis. CONCLUSIONS Our analysis confirms the favorable prognosis of metastatic gastric cancer patients with a defective MMR system and suggests that expression of TILs might also be linked to better outcome. Because of the correlation between defective MMR status and measures of immune system activity, this group of patients would be the best candidates for novel immunotherapy-based therapies.
Collapse
Affiliation(s)
- Riccardo Giampieri
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Elena Maccaroni
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy.
| | | | - Michela Del Prete
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Kalliopi Andrikou
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Luca Faloppi
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Alessandro Bittoni
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Maristella Bianconi
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Marina Scarpelli
- Department of Pathology, Università Politecnica delle Marche, Ancona, Italy
| | - Raffaella Bracci
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, Università di Cagliari, Cagliari, Italy
| | - Stefano Cascinu
- Department of Medical Oncology, Azienda Ospedaliero Universitaria Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
98
|
Zhang M, Dong Y, Liu H, Wang Y, Zhao S, Xuan Q, Wang Y, Zhang Q. The clinicopathological and prognostic significance of PD-L1 expression in gastric cancer: a meta-analysis of 10 studies with 1,901 patients. Sci Rep 2016; 6:37933. [PMID: 27892511 PMCID: PMC5124943 DOI: 10.1038/srep37933] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
The prognostic value of programmed death-ligand 1 (PD-L1) in gastric cancer (GC) remains controversial. To clarify this problem, we performed a meta-analysis of research studies identified in the PubMed, EMBASE and the Cochrane Library databases. A total of 1,901 patients in 10 studies were enrolled in this meta-analysis, and the pooled hazard ratio (HR) of 1.64 (95% CI 1.11 to 2.43; P = 0.01) indicated that PD-L1 expression is associated with a shorter overall survival (OS). The pooled odds ratios (ORs) indicated that PD-L1 expression was associated with tumour size (OR = 1.87, 95% CI 1.25 to 2.78; P = 0.002) and lymph node status (OR = 2.17, 95% CI 1.04 to 4.52; P = 0.04). However, PD-L1 had no correlation with gender, age, cancer location, differentiation, depth of invasion, and tumour stage. This meta-analysis indicates that PD-L1 expression is a valuable predictor of the prognosis of patients with GC. PD-L1 expression could be used for identifying a subgroup of patients, who would potentially benefit from targeted therapy against PD-1 or PD-L1. Well-designed large-cohort studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yuandi Dong
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Haitao Liu
- Department of Intensive Care Unit, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yan Wang
- Department of Medical Oncology, Heilongjiang Provincial Hospital, Harbin 150000, China
| | - Shu Zhao
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Qijia Xuan
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Yan Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| |
Collapse
|
99
|
Solinas C, Pusole G, Demurtas L, Puzzoni M, Mascia R, Morgan G, Giampieri R, Scartozzi M. Tumor infiltrating lymphocytes in gastrointestinal tumors: Controversies and future clinical implications. Crit Rev Oncol Hematol 2016; 110:106-116. [PMID: 28109400 DOI: 10.1016/j.critrevonc.2016.11.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/06/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
Chronic inflammation following infections, autoimmune diseases or exposure to environmental irritants plays a crucial role in tumor development and influences the host immune response to neoplastic cells. The presence of an anti-tumor immune infiltrate is often associated with better outcomes in gastro-intestinal primary cancers, particularly in those with high microsatellite instability (MSI-H). Immunotherapeutic drugs inhibiting the PD-1 and PD-L1 pathway showed promising results in the treatment of these patients in the metastatic setting. The aim of this review is to resume the role tumor infiltrating lymphocytes (TILs) play in gastrointestinal tumors, underlining their potential value as a prognostic and predictive biomarker. TILs assessment could identify subsets of patients with high extent of TILs and better prognosis, that could be spared from adjuvant systemic treatments. Immune infiltration parameters might be additional predictors of a greater benefit from the immunotherapy with the immune checkpoint blockade.
Collapse
Affiliation(s)
- Cinzia Solinas
- Molecular Immunology Unit, Institut Jules Bordet and Université Libre de Bruxelles, Boulevard de Waterloo, 127 1000 Brussels, Belgium.
| | - Grazia Pusole
- Medical Oncology, University of Cagliari, Policlinico Universitario ss 554 bivio Sestu km 4.5, Monserrato, CA, Italy.
| | - Laura Demurtas
- Medical Oncology, University of Cagliari, Policlinico Universitario ss 554 bivio Sestu km 4.5, Monserrato, CA, Italy.
| | - Marco Puzzoni
- Medical Oncology, University of Cagliari, Policlinico Universitario ss 554 bivio Sestu km 4.5, Monserrato, CA, Italy.
| | - Roberta Mascia
- Medical Oncology, University of Cagliari, Policlinico Universitario ss 554 bivio Sestu km 4.5, Monserrato, CA, Italy.
| | | | | | - Mario Scartozzi
- Medical Oncology, University of Cagliari, Policlinico Universitario ss 554 bivio Sestu km 4.5, Monserrato, CA, Italy.
| |
Collapse
|
100
|
Su S, Zou Z, Chen F, Ding N, Du J, Shao J, Li L, Fu Y, Hu B, Yang Y, Sha H, Meng F, Wei J, Huang X, Liu B. CRISPR-Cas9-mediated disruption of PD-1 on human T cells for adoptive cellular therapies of EBV positive gastric cancer. Oncoimmunology 2016; 6:e1249558. [PMID: 28197365 DOI: 10.1080/2162402x.2016.1249558] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/01/2016] [Accepted: 10/13/2016] [Indexed: 12/22/2022] Open
Abstract
The successful use of immune cell checkpoint inhibitors PD-1 and PD-L1, over the past 5 y has raised the concern of using immunotherapy to treat various cancers. Epstein-Barr virus-associated gastric cancer (EBVaGC) exhibits high infiltration of lymphocytes and high amplification of immune-related genes including PD-L1 as distinguished from Epstein-Barr virus-non-associated gastric cancer (EBVnGC). Here, we presume that this PD-1/PD-L1 pathway may hinder the efficacy of adoptive T cell therapy toward EBVaGC. These studies reveal possibility of generating PD-1-disrupted CTL by CRISPR-Cas9 system and demonstrate enhanced immune response of these PD-1-disrupted CTLs to the EBV-LMP2A antigen and superior cytotoxicity to the EBV-positive gastric cancer cell. In addition, when combined with low-dose radiotherapy, these PD-1-disrupted CTLs mediated an impressive antitumor effect in a xenograft mouse model of EBVaGC. Taken together, these studies illustrate PD-1/PD-L1-mediated immune tolerance of EBVaGC and provide a new strategy for targeting immune checkpoints to break the tolerance for the T cell-based adoptive therapy.
Collapse
Affiliation(s)
- Shu Su
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Zhengyun Zou
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Fangjun Chen
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Naiqing Ding
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Juan Du
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Jie Shao
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Lin Li
- Department of Pathology of Drum Tower Hospital, Medical School of Nanjing University , Nanjing, China
| | - Yao Fu
- Department of Pathology of Drum Tower Hospital, Medical School of Nanjing University , Nanjing, China
| | - Bian Hu
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, National Resource Center for Mutant Mice , Nanjing, China
| | - Yang Yang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Huizi Sha
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Fanyan Meng
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Jia Wei
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| | - Xingxu Huang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University, National Resource Center for Mutant Mice, Nanjing, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University , Nanjing, China
| |
Collapse
|